Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Jun 1.
Published in final edited form as: Arch Gen Psychiatry. 2009 Jun;66(6):666–676. doi: 10.1001/archgenpsychiatry.2009.41

β2-Nicotinic Acetylcholine Receptor Availability during Acute and Prolonged Abstinence from Tobacco Smoking

Kelly P Cosgrove 1,2, Jeffery Batis 1,2, Frederic Bois 1,2, Paul K Maciejewski 1, Irina Esterlis 1,2, Tracy Kloczynski 1,2, Stephanie Stiklus 1,2, Suchitra Krishnan-Sarin 1, Stephanie O'Malley 1, Edward Perry 1,2, Gilles Tamagnan 3, John P Seibyl 3, Julie K Staley 1,2
PMCID: PMC2796827  NIHMSID: NIHMS162543  PMID: 19487632

Abstract

Context

β2*-nicotinic acetylcholine receptor (β2*-nAChR) availability is higher in recently abstinent smokers compared to never smokers. Variations in β2*-nAChR availability over the course of abstinence may be related to the urge to smoke, the extent of nicotine withdrawal and successful abstinence.

Objective

To examine changes in β2*-nAChR availability during acute and prolonged abstinence from tobacco smoking and to determine how changes in β2*-nAChR availability were related to clinical features of tobacco smoking.

Design

Tobacco smokers participated in up to 4 [123I]5-IA-85380 ([123I]5-IA) single photon emission computed tomography (SPECT) scans during abstinence: 1 day (n=7), 1 week (n=17), 2 weeks (n=7), 4 weeks (n=11), and 6-12 weeks (n=6). Age-matched nonsmokers participated in 1 [123I]5-IA SPECT scan. All subjects completed 1 magnetic resonance imaging study.

Setting

Academic imaging center.

Participants

Tobacco smokers (n=19) and an age-matched nonsmoker comparison group (n=20).

Main Outcome Measure

[123I]5-IA SPECT images were converted to distribution volume and were analyzed using regions of interest.

Results

Compared to nonsmokers, β2*-nAChR availability in the striatum, cortex and cerebellum of smokers was not different at one day of abstinence, significantly higher at 1week of abstinence and not different at 4 or 6-12 weeks of abstinence. In smokers, at 6-12 weeks of abstinence, β2*-nAChR availability was significantly lower in the cortex and cerebellum compared to 1 week of abstinence. Additionally, cerebellar β2*-nAChR availability at 4 weeks of abstinence was positively correlated with craving on the day of scan.

Conclusions

These data suggest higher β2*-nAChR availability persists up to 1 month of abstinence, and normalizes to nonsmoker levels by 6-12 weeks of abstinence from tobacco smoking. These marked and persistent changes in β2*-nAChR availability may contribute to difficulties with tobacco cessation.


A wealth of evidence from postmortem1-3 and preclinical4-7 studies demonstrate smoking- and nicotine-induced elevations in nicotinic acetylcholine receptors (nAChRs) throughout the brain. Previously, we demonstrated in vivo higher β2-subunit containing nAChR (β2*-nAChR) availability in recently abstinent tobacco smokers compared to nonsmokers8. Higher β2*-nAChR availability in smokers may be due to a variety of molecular changes including increased assembly of α4 and β2 subunits in the endoplasmic reticulum9, increased transport of receptors to the membrane10, decreased receptor turnover11, and/or the presence of nicotine promoting intracellular maturation of the α4β2-nAChR to a high-affinity conformation12. Higher β2*-nAChR availability is thought to functionally reflect higher numbers of desensitized receptors13, 14. Additionally, smoking one cigarette led to greater than 88% receptor occupancy13 suggesting that smokers maintain saturation of β2*-nAChRs over the day. Thus, a chronic tobacco smoker who maintains persistently elevated nicotine levels may experience repeated cycles of nAChR activation and desensitization over each day14.

Preclinical studies have demonstrated that nAChR levels return to control levels after termination of nicotine exposure, but with great variability in the time course15-17. A postmortem human study indicated that individuals who had quit smoking at least 2 months prior to their death (range 2 months-30 years) had nicotine binding levels similar to control nonsmokers2. A recent in vivo study in a small number of male smokers demonstrated a trend for a normalization of the β2-nAChR by 21 days of smoking cessation18. However, these data need to be confirmed in a larger, more heterogeneous population during prolonged abstinence.

The exact subunit combination of nAChR that upregulate in response to nicotine is emerging. NAChRs that contain α4 and β2 subunits 19, 20 are the most abundant nAChRs in brain, and nicotine demonstrates the highest affinity for these receptors [reviewed by 21]. In recent years evidence has emerged that the β2*-nAChRs are a critical neural substrate mediating the effects of nicotine in brain. Much of this information has been derived from studies in β2 knockout mice. These studies have demonstrated that the β2-subunit is critical for self-administration 22, conditioned place preference23, discriminative stimulus and taste aversion24, dopamine release 22, 25, 26, dopamine-dependent locomotor activation27 and enhancement of incentive aspects of motivation28 of nicotine. Studies in wild-type animals further confirm the β2-subunit is critical to the reinforcing properties of nicotine29. The β2-subunit also determines the sensitivity to nicotine30-32, but does not play a critical role in nicotine withdrawal symptoms in rodents33. Importantly, nicotine induced increases in nAChR, termed “upregulation”, are conferred by a specific microdomain that is in the β2-subunit 34 and thus this upregulation is confined to nAChR that contain the β2-subunit4, 19, 31, 35-38.

β2*-nAChR availability can be measured in vivo with [123I]-5-IA-85380 ([123I]5-IA) and single photon emission computed tomography (SPECT). [123I]5-IA is a nicotinic agonist that binds with high affinity to the nicotine binding site on nAChR that contain the β2-subunit39. This ligand demonstrates low nonspecific binding40 and has acceptable dosimetry in human subjects with high brain uptake41, 42 and good test-retest reproducibility43. Because [123I]5-IA is administered at a “trace” dose (<1% occupancy) for SPECT imaging it does not interfere with receptor and cell function. Imaging with [123I]5-IA SPECT in nonhuman primate and human subjects results in a binding pattern that is consistent with the established regional distribution of β2-nAChR and is highest in the thalamus and intermediate throughout the cortex and cerebellum43, 44.

The primary goal of the present study was to evaluate the time course of change in β2*-nAChR availability over prolonged abstinence using [123I]5-IA SPECT. A secondary goal was to explore the relationships between β2*-nAChR availability and behavioral features of tobacco smoking and withdrawal. We hypothesized that during acute abstinence, at 1 day of withdrawal, β2*-nAChR availability would be lower compared to 1 week of abstinence due to the presence of nicotine in the brain, which would block radiotracer binding. Consistent with our previous study8, we hypothesized that compared to control nonsmokers, β2*-nAChR availability would be higher at 1 week of withdrawal, and then would progressively decline over prolonged abstinence.

Methods

Subjects

Nineteen healthy tobacco smokers (9 men, 10 women; 41.1±9.0 years; 13 Caucasian, 5 African American, 1 Hispanic) and 20 age-matched healthy controls (9 men, 11 women; 42.4±9.8 years; 10 Caucasian, 7 African American, 2 Hispanic, 1 Asian) participated in this study. Smokers participated in up to four [123I]5-IA SPECT scans and one magnetic resonance imaging (MRI) study. Subjects were grouped into the following time points of abstinence: 1 day (1.0±0 days; mean ± SD, n=7), 1 week (7.7±1.4 days, n=17), 2 weeks (17.9±3.0 days, n=7), 4 weeks (30.5±4.3 days, n=11), and 6-12 weeks (69.0±23.5 days, n=6). Subjects were grouped as described due to the difficulty in retaining subjects who remained abstinent over long periods and due to the challenges in having subjects complete time consuming (e.g., >8 h day) scans on multiple days. Subjects were scanned during a range of days at each time point due to scheduling constraints. Nonsmoker controls (n=20) participated in one [123I]5-IA SPECT scan and one MRI study.

This study was approved by the Yale University School of Medicine Human Investigation Committee, the West Haven Veterans Administration Human Subjects Subcommittee, and the Radiation Safety Committee. The use of the radiotracer, [123I]5-IA, was approved by the Food and Drug Administration. Subjects were recruited by word of mouth, posters, and newspaper advertisements from the community. Eligibility was determined as follows. All subjects had a medical examination by a study physician to exclude any major medical issues or neurological disorders. This included a physical examination, electrocardiogram, serum chemistries, thyroid function studies, complete blood count, urinalysis, and urine toxicology screening. Subjects were given structured interviews using the Structured Clinical Interview for Diagnostic and Statistical Manual of Mental Disorders (SCID) to rule out any Axis I Disorder except for Nicotine Dependence. All tobacco smokers had to smoke ≥10 cigarettes per day for at least one year. Smoking status was confirmed by plasma cotinine levels >150 ng/mL, urine cotinine levels >100 ng/mL and carbon monoxide levels >11 on the day of intake. Smokers were helped to quit smoking using Clinical Practice Guidelines and contingency management45-47. Briefly, contingency management is a behavioral therapy in which reinforcement is provided contingent upon a successful response. In this study, monetary reinforcement was provided contingent upon abstinence from smoking measured by urine cotinine and breath carbon monoxide levels. Breath carbon monoxide and urine cotinine levels were monitored daily for the first 8 days of smoking cessation and a minimum of twice weekly thereafter. Subjects were instructed that they could not use any form of nicotine replacement therapy or medication throughout the study. All control subjects were nonsmokers (defined as <100 cigarettes in lifetime) and had no history of significant medical illness or major head trauma. Nonsmoking status, was confirmed by plasma cotinine levels <15 ng/mL, urine cotinine levels <100 ng/mL and carbon monoxide levels <11 ppm on the day of intake and day of scan. All women were required to have a negative pregnancy test during the screening process and prior to radiotracer injection on each study day. Plasma nicotine and cotinine levels were measured as previously described8. Urine cotinine levels were measured with either Accutest NicoMeter™ cotinine test strips (Jant Pharmacal, Encino, CA) or NicAlert™ cotinine test strips (Nymox Pharmaceutical, Hasbrouck Heights, NJ).

The severity of nicotine dependence was assessed at intake with the Fagerström Test for Nicotine Dependence (FTND)48, and craving and nicotine withdrawal symptoms were assessed with the Urge to Smoke Questionnaire (QSU)49 and the Minnesota Nicotine Withdrawal Scale (MNWS)50, respectively, at baseline, e.g., on the day of their intake prior to quitting smoking, and on each day of their participation in [123I]5-IA scans. The QSU has two main factors, the intention/desire to smoke (QSU-Intent) and relief of negative affect and withdrawal (QSU-Relief).

MR Imaging

MRI studies were obtained on a 1.5 Tesla Siemens camera in a standard orientation (TE=5-7 ms; TR=24ms; a 256×192 matrix; 1 NEX; FOV 30 cm; 124 contiguous slices with 1.2 mm thickness) and were used for coregistration to the SPECT images to provide an anatomical guide for placement of regions of interest.

[123I]5-IA SPECT scan

All subjects received a 0.6-g saturated solution of potassium iodide, to protect their thyroid from possible exposure to radioactive iodide, in the hour prior to radiotracer administration. [123I]5-IA was synthesized as previously described 51 and administered as a bolus to constant infusion at a ratio of 7.0 for 8 hours. Subjects were injected with equivalent doses of a bolus (mean±SD, MBq) and constant infusion (mean±SD, MBq/h) as follows: control nonsmokers (157.9±14.6 MBq, 22.8±2.1 MBq/h), 1 day abstinent smokers (134.5±24.6 MBq, 20.5±3.8 MBq/h), 1 week abstinent smokers (154.3±15.9 MBq, 22.2±2.6 MBq/h), 2 week abstinent smokers (159.4±5.6 MBq, 23.3±0 MBq/h), 4 week abstinent smokers (158.9±7.4 MBq, 22.6±1.7 MBq/h) and 6-12 week abstinent smokers (141.5±30.5 MBq, 21.9±3.5 MBq/h). Three consecutive 30-min emission scans and one 15-min simultaneous transmission and emission protocol scan (STEP) were obtained between hours 6 and 8 of the [123I]5-IA infusion on a Picker PRISM 3000 XP (Cleveland, OH) SPECT camera. The PRISM 3000 XP is a 3-headed camera equipped with a low energy, ultra-high resolution fanbeam collimator (photopeak window, 159 keV ± 10%; matrix 128×128) with a uniform sensitivity across the field of view. A 57Co-distributed source was measured with each experiment to control for day-to-day variation in camera sensitivity. The axial resolution (full width at half maximum) is 12.2 mm, measured with a 123I line source in water in a cylindrical phantom. Blood was drawn prior to injection and at the beginning and end of the emission scans for analysis of plasma total parent and free fraction of parent tracer in plasma (fP, free fraction). The chemical fate of [123I]5-IA post injection was assessed in plasma as previously described51. Briefly, plasma total parent was assessed by acetonitrile protein denaturation, while free fraction was determined by ultrafiltration using Centrifree units.

Image analysis and outcome measures

Images were reconstructed and analyzed as previously described including a nonuniform attenuation correction 43 with one exception. Specifically, in subjects who had more than one SPECT scan, the second and subsequent SPECT scans were coregistered to the same position as the first scan in order to apply the same region of interest template for that subject. MRIs were coregistered to the SPECT images to provide an anatomical guide for placement of the regions of interest using Medx (version 3.4) software (Medical Numerics, Inc., MD). Regions of interest chosen were those known to contain β2-nAChRs and included frontal, parietal, anterior cingulate, temporal and occipital cortices, thalamus, striatum (an average of caudate and putamen) and cerebellum. Regions-of-interest are corrected to account for differences in size. Two raters conducted the analysis. Variability between raters was <12 % across regions-of-interest. The mean of the analysis from the two raters is reported.

The outcome measure VT/fP (regional activity divided by free plasma parent between 6 and 8 hours), was used to correct for possible differences in radiotracer metabolism or plasma protein binding between groups and subjects. Specifically, VT/fP equals [123I]5-IA uptake in a region-of-interest (kBq/cc) / free plasma parent (kBq/mL)52. We refer to VT/fP as β2*-nAChR availability because we are only measuring receptors that are “available” to be bound by radiotracer. Receptors that are already occupied, e.g., by residual nicotine, a pharmacologically active metabolite (cotinine or nornicotine) or by endogenous neurotransmitter (acetylcholine) are not available. VT/fP is proportional to the binding potential (BP, mL/g=Bmax/KD), which is proportional to the receptor number (Bmax) at equilibrium, given the assumptions that there is no change in affinity (KD) and that nondisplaceable (nonspecific and free) uptake does not differ between subjects or comparison groups. As described previously43, there is no appropriate reference region for this radiotracer, so nondisplaceable [123I]5-IA uptake could not be measured. The measures of total plasma parent (kBq/mL), fP and free plasma parent (kBq/mL) which is defined as total parent * fP were compared between groups to determine differences in radiotracer metabolism or protein binding.

Statistical Analyses

Data were analyzed using SAS version 9.1 (SAS Institute Inc, Cary, NC). Differences in blood measures (total and free parent, and fP) and regional β2*-nAChR availability, VT/fP, between subjects in the nonsmoker control group and each of the 1 day, 1 week, 2 week, 4 week, and 6-12 week abstinent smoker groups were assessed using two-sample t-tests. Differences in regional brain VT/fP and behavioral measures of tobacco smoking and withdrawal between subjects in the abstinent smoker groups were assessed using repeated measures, mixed-effects regression models with group as a fixed effect and compound symmetry covariance structure across repeated measurements. These between group differences were evaluated using repeated measures, mixed-effects models in order to account for the observations between abstinent smoker groups that were not entirely independent, given that some abstinent smokers contribute observations to more than one group. For models examining between-group differences for which there was a significant effect of group, four planned post hoc between-group comparisons were conducted, i.e., between 1 week abstinent smokers and 1 day, 2 week, 4 week, and 6-12 week abstinent smokers. For these post hoc tests, p-values were Bonferroni corrected for multiple comparisons, and statistical significance was considered at p≤0.00625. Correlational analyses for the associations between receptor availability and smoking assessments were conducted using SPSS version 16.0 (SPSS Inc. Headquarters, Chicago, IL). Correlations between β2*-nAChR availability, VT/fP, at each time point and clinical variables (smoking, craving, withdrawal) were assessed with Spearman's rho correlation coefficients. Different slopes for each abstinence time point were estimated to illustrate the relationship between significant clinical variables and β2*-nAChR availability. Due to multiple comparisons, statistical significance was considered at p≤0.01.

Results

Clinical population

Nineteen tobacco smokers and 20 age-matched nonsmokers were included in the study. Smokers who participated in scans at different times since the last cigarette were equivalent in age, level of nicotine dependence (as assessed by the FTND at intake), cigarettes smoked per day, and years of smoking (Table 1). Plasma cotinine and nicotine levels and carbon monoxide measurements were negligible in nonsmokers and in smokers were highest at 1 day of abstinence and decreased over time, confirming abstinence from smoking (Table 1).

Table 1. Demographics and Smoking Characteristics of Nonsmoker Controls and Smokers during Acute and Prolonged Abstinence.

Characteristics Nonsmoker
Control
(n=20)
Smoker
1 day
(n=7)
Smoker
1 week
(n=17)
Smoker
2 weeks
(n=7)
Smoker
4 weeks
(n=11)
Smoker
6-12 weeks
(n=6)
Age, years 42.4±9.8 42.7±8.2 41.7±9.4 43.4±11.7 43.9±7.5 38.7±7.0
FTND 0 5.9±2.8 5.5±2.6 5.4+2.8 4.9±2.6 6.8±2.3
Cigarettes/Day 0 19.9±10.2 19.7±8.5 22.7±7.9 14.7±3.6 20.3±10.3
Years Smoked 0 21.8±6.3 19.9±7.6 22.0±7.4 20.8±7.7 21.2±6.6
Plasma Cotinine (ng/mL) <15 370.3±185.6 21.1±46.3 <15 <15 <15
Plasma Nicotine (ng/mL) <4 3.6±5.2 <4 <4 <4 <4
Carbon Monoxide 2.2±3.1 12.0±7.2 3.2±2.4 2.9±1.9 2.9±2.2 4.2±3.3

Data are presented as mean ± SD.

β2*-nAChR availability during acute and prolonged abstinence

There were no differences between groups in injected dose, bolus to infusion ratio, or time of scan (data not shown). Concentrations of [123I]5-IA activity in the blood (kBq/mL) were measured in order to correct for potential differences between groups in radiotracer metabolism or protein binding. There were significant differences in total parent (kBq/mL) between nonsmokers and 1 day abstinent smokers and in fP between nonsmokers and 2 week abstinent smokers (Table 2). There were no differences in total parent (kBq/mL) or fP between groups at other time points, or between groups at any time point in free parent (kBq/mL). There was also variability between smokers who participated in multiple scans in changes in β2*-nAChR availability over time with some abstinent smokers showing dramatic changes in β2*-nAChR availability (up to 48% change in the cortex) and others showing barely any difference, e.g., less than 5%, over time (individual data not shown).

Table 2. Blood [123I]5-IA Measures of Nonsmoker Controls and Smokers during Acute and Prolonged Abstinence.

Measure Nonsmoker
Control
(n=20)
Smoker
1 day
(n=7)
Smoker
1 week
(n=17)
Smoker
2 weeks
(n=7)
Smoker
4 weeks
(n=11)
Smoker
6-12 weeks
(n=6)
Total Parent (kBq/mL) 0.333±0.103 0.236±0.032* 0.302±0.081 0.310±0.088 0.305±0.089 0.281±0.088
Free Parent (kBq/mL) 0.112±0.036 0.080±0.018 0.108±0.032 0.123±0.043 0.100±0.024 0.101±0.045
fP 0.342±0.039 0.334±0.073 0.358±0.049 0.398±0.056* 0.337±0.052 0.356±0.052

Data are presented as mean ± SD.

*

Indicates significantly different after Bonferroni's correction from nonsmoker controls.

Regional β2*-nAChR availability, reflected by VT/fP, was compared between nonsmokers and each of the abstinent smoker groups and between the abstinent smoker groups (Table 3 and Figures 1 and 2). In 1 day abstinent smokers as compared to nonsmokers, VT/fP was significantly reduced in the thalamus. In 1 week abstinent smokers as compared to nonsmokers, VT/fP was significantly higher in the striatum, cerebellum and throughout the cortex. In 4 week abstinent smokers as compared to nonsmokers, VT/fP was significantly higher in the occipital cortex (Table 3 and Figures 1 and 2).

Table 3. β2*-nAChR availability (VT/fP) throughout the Brain in Nonsmoker Controls and Smokers during Acute and Prolonged Abstinence.

Nonsmoker Smoker 1 day Smoker 1 week Smoker 2 weeks Smoker 4 weeks Smoker 6-12 weeks

Region VT/fP* VT/fP % Diff** P VT/fP % Diff P VT/fP % Diff P VT/fP % Diff P VT/fP % Diff P
Thalamus 130.6±22.2 96.9±24.4 -26% .002 138.5±25.9 +6% .322 139.2±35.6 +7% .461 133.8±19.3 +2% .694 115.2±19.7 -12% .140
Striatum 70.0±11.0 66.5±13.1 -5% .492 85.1±16.8 +22% .002 80.9±18.2 +16% .070 79.5±12.6 +14% .037 71.7±13.3 +2% .760
Parietal Cortex 46.0±7.8 44.4±7.5 -3% .659 56.6±9.9 +23% .001 56.4±16.2 +23% .144 52.3±10.9 +14% .071 44.5±9.6 -3% .714
Frontal Cortex 51.0±8.6 47.9±10.8 -6% .450 61.8±13.0 +21% .005 60.9±15.9 +19% .158 58.3±13.8 +14% .078 49.1±12.9 -4% .685
Anterior Cingulate 52.5±7.8 54.8±7.7 +4% .511 67.5±12.2 +29% <.001 63.9±15.1 +22% .098 61.4±10.0 +17% .010 55.3±11.6 +5% .500
Temporal Cortex 57.7±8.7 57.4±10.4 -1% .941 70.2±12.8 +22% .001 67.5±15.3 +17% .048 66.1±9.7 +15% .021 58.2±11.6 +1% .908
Occipital Cortex 54.2±7.7 56.3±11.7 -4% .587 68.6±13.4 +27% .001 66.3±14.3 +22% .067 64.1±10.3 +18% .005 55.5±13.9 +2% .771
Cerebellum 62.9±8.5 59.5±18.4 -5% .649 77.9±15.5 +24% .002 74.0±17.0 +18% .143 73.2±11.5 +16% .009 60.6±13.5 -4% .605
*

VT/fP equals regional [123I]5-IA uptake (kBq/cc)/free plasma parent (kBq/mL);

**

Percent Difference from Nonsmokers = ((Smoker-Nonsmoker)/Nonsmoker)*100;

p-values <0.00625 are significant after Bonferroni's correction

Figure 1.

Figure 1

β2*-nAChR availability (VT/fP) is shown in individual nonsmokers (open diamonds) and tobacco smokers (filled circles) at 1 day, 1 week, 2 weeks, 4 weeks, and 6-12 weeks of abstinence in the thalamus, striatum (average of caudate and putamen), cortex (average of cortical regions including parietal, frontal, anterior cingulate, temporoinsular, and occipital cortex), and cerebellum. The line in each scatter plot represents the mean value of those subjects. * indicates significant difference from control nonsmokers after Bonferroni's correction using two-sample t-tests. † indicates significant difference from 1 week abstinent smokers after Bonferroni's correction using planned post-hoc between-group comparisons subsequent to the analysis of repeated measures mixed-effects regression models including the overall effect of abstinent smoker group.

Figure 2.

Figure 2

Mean parametric images illustrating β2*-nAChR availability in nonsmokers and tobacco smokers at 1 day, 1 week, 2 weeks, 4 weeks, and 6-12 weeks of abstinence at similar transaxial levels of brain. The color scale is shown with red, yellow, green and blue corresponding to VT/fP values.

Among the abstinent smoker groups, there were significant between group differences in VT/fP in the thalamus (F[4,25]=4.12, p=0.011), parietal cortex (F[4,25]=2.95, p=0.040), frontal cortex (F[4,25]=2.88, p=0.043), anterior cingulate (F[4,25]=3.75, p=0.016), occipital cortex (F[4,25]=3.42, p=0.023), and cerebellum (F[[4,25]=4.00, p=0.012). Compared to 1 week abstinent smokers, 1 day abstinent smokers had significantly lower VT/fP in the thalamus (t[25]=-3.51, corrected p=0.007) and cerebellum (t[25]=-3.02, corrected p=0.023). Compared to 1 week abstinent smokers, 6-12 week abstinent smokers had significantly lower VT/fP in the parietal cortex (t[25]=-2.87, corrected p=0.033), frontal cortex (t[25]=-2.80, corrected p=0.039), anterior cingulate (t[25]=-3.21, corrected p=0.014), occipital cortex (t[25]=-3.10, corrected p=0.019) and cerebellum (t[25]=-3.17, corrected p=0.016). There were no significant differences in VT/fP between 1 week abstinent smokers compared to smokers at 2 or 4 weeks of abstinence (Table 3 and Figures 1 and 2).

Relationship between β2*-nAChR availability and clinical features

There were significant differences in QSU-Intent (F[4,25]=7.41, p<0.015) and QSU Relief (F[4,25]=5.63, p=0.002), but not in MNWS scores (F[4,25]=0.33, p=0.857), between groups of abstinent smokers (Table 4). Specifically, overall group differences in QSU-Intent and QSU-Relief were attributable to significantly higher levels of each of these measures on the day of the scan in the 1 day abstinent smoker group as compared to smokers at other time points.

Table 4. Clinical Characteristics of Smokers during Acute and Prolonged Abstinence.

Measure Smoker
1 day
(n=7)
Smoker
1 week
(n=17)
Smoker
2 weeks
(n=7)
Smoker
4 weeks
(n=11)
Smoker
6-12 weeks
(n=6)
MNWS 11.2±3.2 10.5±2.5 11.7±3.2 12.8±2.8 12.6±3.3
QSU Intent 26.0±2.7* 16.3±2.1 16.0±2.7 12.3±2.3 14.8±2.8
QSU Relief 20.1±2.4* 10.6±1.8 10.4±2.4 10.2±2.0 8.3±2.5

Data are presented as least-squares mean ± SE.

*

Indicates significantly different after Bonferroni's correction compared to smokers at other time points.

There were significant correlations between regional β2*-nAChR availability and clinical features and assessment scores at baseline, e.g., at intake prior to quitting smoking (Table 5). Specifically, baseline QSU-Intent scores correlated negatively with β2*-nAChR availability at 1 day of abstinence in the thalamus (rho=-0.90, p=0.006) and parietal cortex (rho=-0.88, p=0.008). There were also significant correlations between regional β2*-nAChR availability and assessment scores taken at each abstinent time point (Table 6). Specifically, a positive correlation was observed between cerebellar β2*-nAChR availability and craving on both the QSU-Intent (rho=0.74, p=0.01) and QSU-Relief (rho=0.74, p=0.01) at 4 weeks of abstinence. There were no significant correlations between baseline smoking variables, e.g., FTND, number of years smoked, or number of cigarettes smoked per day with β2*-nAChR availability at any time point (data not shown).

Table 5. Correlations between β2*-nAChR Availability and Baseline Assessments.

Smoker 1 day (n=7) Smoker 1 week (n=10)** Smoker 2 weeks (n=7) Smoker 4 weeks (n=11) Smoker 6-12 weeks (n=6)

Region MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief
Thalamus .16 -.90* -.56 .06 .29 .17 -.31 - - .22 -.34 -.05 .83 - -
Striatum -.05 -.76 -.56 -.01 .16 .17 -.31 - - .17 -.29 -.29 .43 - -
Parietal Cortex .27 -.88* -.56 -.15 .45 -.20 .02 - - -.03 -.49 -.67 .66 - -
Frontal Cortex .09 -.78 -.49 .06 -.31 -.18 -.31 - - .08 -.53 -.43 .89 - -
Anterior Cingulate -.22 -.63 -.51 .02 .46 .18 -.20 - - .17 -.35 -.39 .77 - -
Temporal Cortex -.34 -.67 -.63 .07 .46 .23 -.34 - - .25 -.37 -.25 .89 - -
Occipital Cortex -.11 -.67 -.32 .22 .12 .59 -.45 - - .48 -.73 .05 .89 - -
Cerebellum -.02 -.85 -.52 .11 -.09 .12 -.34 - - .64 -.61 .21 .77 - -
*

p≤0.01 (two-tailed)

**

Due to missing QSU baseline data, correlations were only conducted in 10 of 17 subjects.

Due to missing QSU baseline data, Spearman's correlations could not be conducted.

Table 6. Correlations between β2*-nAChR Availability and Assessments on Day of Scan.

Smoker 1 day (n=7) Smoker 1 week (n=17) Smoker 2 weeks (n=7) Smoker 4 weeks (n=11) Smoker 6-12 weeks (n=6)

Region MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief MNWS QSU Intent QSU Relief
Thalamus -.72 -.14 -.52 -.12 .21 -.03 -.18 .06 -.26 -.06 .36 .54 .09 .44 .40
Striatum -.52 .18 -.67 -.31 .03 -.15 -.18 0.6 -.26 -.41 .15 .32 .03 .79 .39
Parietal Cortex -.45 .14 -.36 -.28 -.10 -.36 .25 .46 .10 -.60 -.19 .23 .03 .18 .13
Frontal Cortex -.52 .00 -.36 -.36 .04 -.17 -.18 .06 -.26 -.63 -.16 .26 -.03 .44 .39
Anterior Cingulate -.31 .21 -.60 -.01 .01 -.08 .07 .11 -.12 -.34 .10 .19 -.09 .56 .39
Temporal Cortex -.56 .00 -.74 .04 .12 -.08 -.04 .11 -.12 -.33 .18 .34 -.03 .44 .39
Occipital Cortex -.38 -.25 -.34 .01 .16 .08 -.07 .00 -.02 -.25 .33 .41 -.03 .44 .39
Cerebellum -.74 -.32 -.61 -.23 .12 -.05 -.07 -.27 .04 .14 .74* .74* -.09 .56 .39
*

p≤0.01 (two-tailed)

Comment

The present study examined the time course of changes in β2*-nAChR availability during acute and prolonged abstinence in tobacco smokers compared to nonsmokers using [123I]5-IA SPECT. The present findings demonstrate higher β2*-nAChR availability in the striatum, cerebellum and cerebral cortex in tobacco smokers at 1 week of abstinence compared to nonsmokers, but similar or lower β2*-nAChR availability to nonsmokers smokers at 1 day and 6-12 weeks of abstinence. While there is not a significant difference between β2*-nAChR availability in smokers at 2 and 4 weeks of abstinence compared to nonsmokers, there remains a robust difference, e.g., higher β2*-nAChR availability in smokers at 2 weeks (16-23%) and 4 weeks (14-18%) of abstinence in the striatum, cerebellum and cortex compared to nonsmokers, that does not return to nonsmoker levels until 6-12 weeks of abstinence (-4-5% difference). There are two primary implications to these results. First, at 1 day of abstinence there is still residual nicotine, or a pharmacologically active metabolite of nicotine, such as cotinine or nornicotine present in the brain that interferes with radiotracer binding, thus leading to the appearance of lower β2*-nAChR availability. Second, the normalization of the β2*-nAChR is prolonged, requiring up to 6-12 weeks of abstinence to fully return to nonsmoker levels.

Interestingly, in the smokers at 1 day of abstinence, the levels of total parent of the radiotracer were significantly lower, but normalized quickly, by 1 week of abstinence. This highlights the impact of nicotine or another chemical in tobacco smoke on metabolism, e.g., because nicotine was still present, it may have changed the metabolism of the radiotracer, resulting in lower total parent at 1 day of abstinence. Cytochrome P450 (CYP 2A6) is primarily responsible for the metabolism of nicotine to its main metabolite cotinine53. There is evidence that nicotine is metabolized faster in smokers than in nonsmokers and there are genetically-mediated differences in the metabolism of nicotine in smokers54. Additionally, nicotine can interfere with metabolism of other drugs55. Consistently, we expect that [123I]5-IA is metabolized in the liver by enzymes in the cytochrome P450 family, such as CYP2A6, which acts on nicotine, and CYP2B6 and CYP2D6, which catalyze the dealkylation of aromatic ethers56, 57. In radiotracer imaging studies it is imperative that brain uptake is corrected for radiotracer metabolism, since differences in metabolism of the radiotracer determine how much radiotracer is available to the brain, e.g., fast metabolizers will have less radiotracer available to brain, while slow metabolizers will have more radiotracer available to brain for a given dose. By using the outcome measure VT/fP, we correct for individual differences in radiotracer metabolism and protein binding.

In the present study we report a negative correlation between baseline craving scores, e.g., prior to quitting smoking, and β2*-nAChR availability at 1 day of abstinence in the thalamus and parietal cortices. Because receptor availability is defined as receptors that are available to be bound by the radiotracer, at 1 day of abstinence subjects with lower receptor availability have more nicotine present in the brain occupying receptors and blocking the radiotracer from binding to the receptor. Thus, we believe that subjects who reported high baseline craving, likely smoked more cigarettes immediately prior to their quit day and had lower receptor availability at 1 day of abstinence. However, the experience of craving in the presence of nicotine occupancy of the β2*-nAChR is not unusual. Smokers experience craving within 2 h of their last cigarette despite continued occupancy of the receptor by nicotine13. Dopamine release has been associated with the feeling of craving58, and both nicotine59 and cotinine60 have been shown to facilitate dopamine release, thus the prolonged partial occupancy of the receptor by nicotine or cotinine, may contribute to the feelings of craving that are reported 2 h after the last cigarette and throughout the first week of abstinence.

We also report a positive correlation between craving on the day of scan at 4 weeks of abstinence and cerebellar β2*-nAChR availability at 4 weeks of abstinence. Thus, those individuals with higher cerebellar β2*-nAChR availability at 4 weeks of abstinence report a greater urge to smoke on that day. Associations between nicotine and craving have previously been identified in the thalamus61, 62 and generally in areas associated with emotion and reward, and those with high densities of nAChRs (see63 for review). Interestingly, a previous study found associations between craving and regions subserving motor functions including the primary motor cortex, premotor cortex and supplementary motor area64 which require input from the cerebellum. Additionally, when smokers are told to actively resist craving during cigarette cue exposure, the motor cortex is deactivated65. Our finding of a link between the cerebellum and craving further suggests that craving has a motor component, so that craving for cigarettes may elicit preparation for action or voluntary movement, goal directed actions (e.g., lighting a cigarette, bringing a cigarette up to the mouth) and/or motor imagery which are linked to the motor system66, 67. It can be estimated that over the course of 20 years, a one pack per day smoker (assuming 11 puffs on average per cigarette68) may perform the action of bringing a lit cigarette to the mouth over 1.5 million times. Thus, the physical action of smoking a cigarette is likely to be critically tied to craving during abstinence.

In addition to the role of the cerebellum in motor functions, there is increasing interest in the involvement of the cerebellum in cognition. Specifically, activation of the cerebellum has been associated with tasks requiring explicit, episodic memory, e.g., recall of autobiographical events69, 70. With regard to drug abuse, imaging studies have determined that the cerebellum is activated in response to smoking-related cues71, and is associated with cue-induced craving in cocaine abusers72, 73 and recently abstinent alcoholics74. Activity in the cerebellum has also been linked to executive dysfunction in cocaine users75. Taken together, these studies and the present findings highlight the cerebellum as a brain region that is critically linked to craving by both motor and cognitive functions.

We report no significant correlations between nicotine withdrawal and β2*-nAChR availability. This is consistent with the preclinical literature suggesting that the β2*-subtype does not play a critical role in the physical symptoms of nicotine withdrawal33, 76. In this study, subjects reported a mild-moderate level of nicotine withdrawal symptoms at baseline and over the course of the study, thus these results require replication in a larger sample with a greater range of nicotine withdrawal symptoms. Additionally, we did not obtain significant correlations between β2*-nAChR availability at 1 week of abstinence and clinical features. We previously found a negative correlation between the urge to smoke to relieve withdrawal symptoms and β2*-nAChR availability in the sensorimotor cortex at approximately 1 week of abstinence8. This discrepancy may be due to differences in correlational analysis methods, e.g., voxel-based analyses in the previous study versus spearman's rho correlations with regions-of-interest in the current study. Voxel-based analyses may be more sensitive to detecting significance in smaller brain regions, but that was beyond the scope of the current study. The lack of additional correlations at 1 week of abstinence was previously discussed8.

Consistent with our previous study8 we report significantly higher β2*-nAChR availability in smokers at one week of abstinence in the cortex, striatum and cerebellum, but not thalamus compared to nonsmokers. The difference between recently abstinent smokers and never smokers in the previous study8 was of a greater magnitude, e.g., 26-36% in the cerebral cortex and 27% in the striatum, than in the current study, e.g., 21-29% in the cerebral cortex and 22% in the striatum. This may be due to the older average age (approximately 5 years) of subjects in the current study, since β2*-nAChR availability has been shown to decrease with age in nonsmokers77. This study and previous in vivo PET78 and SPECT8, 18 studies report no upregulation of thalamic β2*-nAChR availability during acute abstinence, which conflicts with postmortem2 and animal8, 79 studies. In general, this may be due to differences in methodology or the higher relative dose of nicotine in the postmortem and animal studies. However, two smokers in the current study exhibited increased β2*-nAChR availability in the thalamus compared to nonsmokers, highlighting the role of individual differences in receptor regulation.

This study is also consistent with a previous study18 demonstrating that higher β2*-nAChR availability in recently abstinent tobacco smokers compared to nonsmokers is temporary. In the previous study in men, β2*-nAChR availability decreased to nonsmoker levels in some subjects by 21 days of abstinence. Specifically, compared to nonsmokers, smokers had significantly lower β2*-nAChR availability at 4 hr of abstinence, significantly higher β2*-nAChR availability at 10 days of abstinence, and similar β2*-nAChR availability at 21 days of abstinence. Additionally, they reported significantly lower β2*-nAChR availability at 21 days of abstinence compared to 10 days of abstinence. One difference is that the previous study18 had significantly lower β2*-nAChR availability in all regions at 4 hr of abstinence compared to the nonsmokers, while the current study reports lower thalamic but similar β2*-nAChR availability in the striatum, cerebellum and throughout the cortex compared to the nonsmoker group at 1 day of abstinence. This is interesting and likely due to high levels of residual nicotine or metabolites present in the brain at 4 hr of abstinence (versus ∼24 hours in the current study) resulting in lower β2*-nAChR availability. Also, while the current study did not find a significant decrease, on average, in β2*-nAChR availability by 4 weeks of abstinence compared to 1 week of abstinence, in some subjects normalization did occur by this point. The high heterogeneity of the subject population in the current study compared to the previous study in men only18 likely accounts for the high individual variability with regard to receptor changes during prolonged abstinence. Thus, the present study contributes to the literature with a larger, more heterogeneous subject group, a more prolonged period of abstinence, and additional assessments of behavioral features of tobacco smoking.

Previous preclinical studies demonstrated that in chronically nicotine treated animals, nAChR levels returned to levels observed in control animals after termination of nicotine, but with variable timing ranging between 1 and 3 weeks15, 80, 81. The previous human study18 indicated a return to control levels within 21 days of abstinence; and, the current study indicates that on average, β2*-nAChR availability in recently abstinent tobacco smokers does not normalize until between 4-12 weeks, although, this is highly variable between individuals. The differences in the time course changes may be due to differences in dosing regimen, chronicity of nicotine, route of administration, metabolism between species, specificity of the radioligand, or sex and/or genetic differences in nAChR subunit expression and composition of nicotinic agonist binding sites, but taken together the results consistently highlight a return to control levels or “normalization” of the β2*-nAChR after termination of chronic nicotine. The preclinical studies support a process of prolonged normalization when we consider that 1-3 weeks is substantial in the life span of a rodent. Additionally, this prolonged normalization of the receptor is in line with the protracted withdrawal symptoms reported by tobacco smokers. For example, while withdrawal symptoms such as anger, anxiety, depression and difficulty concentrating tend to peak within the first week of quitting smoking they continue up to 4 weeks after the quit attempt82. We are currently examining variables that may be associated with the rate of normalization.

One limitation of this study is that [123I]5-IA measures the availability of the β2-subunit of the nAChR, primarily the α4β2-subunit, but other subunits also contribute to the regulation of the nAChR by tobacco smoking. Notably, the β2-subunit has been linked primarily to the reinforcing effects of nicotine22, 83, 84 and combines with the α3-6 subunits. Importantly, different β2*-subunit combinations appear to be differentially regulated by nicotine, with α36 and α6α4β2* and α5α4β2* not upregulating or decreasing in response to nicotine38, 85-88 while α6β2 (nonα4) nAChR increase in response to nicotine88. Notably, nAChR subunit expression varies regionally, and differential regulation of these distinct subunit combinations that are measured by a single nicotinic agonist ligand such as [123I]5-IA will result in regional differences in the degree of upregulation in smokers and the degree of receptor normalization during abstinence. Thus, we are likely measuring the β2-subunit in combination with α3-6 subunits leaving the in vivo measurement of the β3-subunit and α7-subunit for the future, with further radiotracer development. Both the α7- and β3-subunit have been implicated in modulating dopamine release14, 89 and thus, may also play a role in the rewarding properties of tobacco smoke. While the α7-subunit does not upregulate in response to chronic nicotine31, it is critically involved along with the β2-subunit in mediating desensitization/inactivation of the neuronal nAChRs in response to chronic nicotine90.

In summary, our data extend the findings of previous studies8, 18 which showed that the upregulation of the β2*-nAChR in recently abstinent tobacco smokers was temporary and could be measured with [123I]5-IA SPECT imaging. Specifically, we demonstrate that the normalization of upregulated β2*-nAChRs in tobacco smokers during smoking cessation is prolonged. This is consistent with the clinical course of tobacco smoking in which craving, withdrawal symptoms, and risk for relapse are prolonged. The variation between individuals in the magnitude of upregulation and rates of normalization may ultimately be used to delineate subgroups based on genetics, sex, and comorbid mental illness and thus to target treatment medications.

Acknowledgments

The authors gratefully acknowledge Dr. Marina Picciotto and Dr. Robert Makuch for helpful comments on the manuscript and Louis Amici and the nuclear technologists at the Institute for Neurodegenerative disorders for technical assistance. We thank the laboratory of Peter Jatlow, MD, for determining plasma nicotine and cotinine levels. The authors have no conflicts of interest related to the manuscript.

Funding: This research was supported in part by National Institute of Health grants RO1 DA015577 and KO2 DA21863 (Staley), KO1 DA20651 (Cosgrove), P50 DA13334 and P50 AA15632 (O'Malley), K25 NS044316 (Maciejewski), and T32 14276. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute on Alcohol Abuse and Alcoholism, the National Institute on Drug Abuse, or the National Institutes of Health.

References

  • 1.Benwell M, Balfour D, Anderson J. Evidence that tobacco smoking increases the density of (-)-[3H]nicotine binding site in human brain. J Neurochem. 1988;50:1243–1247. doi: 10.1111/j.1471-4159.1988.tb10600.x. [DOI] [PubMed] [Google Scholar]
  • 2.Breese C, Marks M, Logel J, et al. Effect of smoking history on [3H]nicotine binding in human postmortem brain. J Pharmacol Exp Ther. 1997;282:7–13. [PubMed] [Google Scholar]
  • 3.Perry D, Dávila-García M, Stockmeier C, Kellar K. Increased nicotinic receptors in brains from smokers: Membrane binding and autoradiography studies. J Pharmacol Exp Therap. 1999;289:1545–1552. [PubMed] [Google Scholar]
  • 4.Nguyen HN, Rasmussen BA, Perry DC. Subtype-selective up-regulation by chronic nicotine of high-affinity nicotinic receptors in rat brain demonstrated by receptor autoradiography. J Pharmacol Exp Ther. 2003;307:1090–7. doi: 10.1124/jpet.103.056408. [DOI] [PubMed] [Google Scholar]
  • 5.Schwartz R, Kellar K. Nicotinic cholinergic receptor binding sites in the brain: regulation in vivo. Science. 1983;220:214–216. doi: 10.1126/science.6828889. [DOI] [PubMed] [Google Scholar]
  • 6.Abreu-Villaca Y, Seidler F, Qiao D, et al. Short-term adolescent nicotine exposure has immediate and persistent effects on cholinergic systems: critical periods, patterns of exposure, dose thresholds. Neuropsychopharmacology. 2003;28:1935–1949. doi: 10.1038/sj.npp.1300221. [DOI] [PubMed] [Google Scholar]
  • 7.Nashmi R, Xiao C, Deshpande P, et al. Chronic nicotine cell specifically upregulates functional alpha 4* nicotinic receptors: basis for both tolerance in midbrain and enhanced long-term potentiation in perforant path. J Neurosci. 2007;27:8202–18. doi: 10.1523/JNEUROSCI.2199-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Staley JK, Krishnan-Sarin S, Cosgrove KP, et al. Human tobacco smokers in early abstinence have higher levels of beta2* nicotinic acetylcholine receptors than nonsmokers. J Neurosci. 2006;26:8707–14. doi: 10.1523/JNEUROSCI.0546-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nashmi R, Dickinson ME, McKinney S, et al. Assembly of alpha4beta2 nicotinic acetylcholine receptors assessed with functional fluorescently labeled subunits: effects of localization, trafficking, and nicotine-induced upregulation in clonal mammalian cells and in cultured midbrain neurons. J Neurosci. 2003;23:11554–67. doi: 10.1523/JNEUROSCI.23-37-11554.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Harkness PC, Millar NS. Changes in conformation and subcellular distribution of alpha4beta2 nicotinic acetylcholine receptors revealed by chronic nicotine treatment and expression of subunit chimeras. J Neurosci. 2002;22:10172–81. doi: 10.1523/JNEUROSCI.22-23-10172.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Peng X, Gerzanich V, Anand R, Whiting P, Lindstrom J. Nicotine-induced increase in neuronal nicotinic receptor results from a decrease in the rate of receptor turnover. Mol Pharmacol. 1994;46:523–530. [PubMed] [Google Scholar]
  • 12.Sallette J, Pons S, Devillers-Thiery A, et al. Nicotine upregulates its own receptors through enhanced intracellular maturation. Neuron. 2005;46:595–607. doi: 10.1016/j.neuron.2005.03.029. [DOI] [PubMed] [Google Scholar]
  • 13.Brody AL, Mandelkern MA, London ED, et al. Cigarette smoking saturates brain alpha 4 beta 2 nicotinic acetylcholine receptors. Arch Gen Psychiatry. 2006;63:907–15. doi: 10.1001/archpsyc.63.8.907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Picciotto MR, Addy NA, Mineur YS, Brunzell DH. It is not “either/or”: Activation and desensitization of nicotinic acetylcholine receptors both contribute to behaviors related to nicotine addiction and mood. Prog Neurobiol. 2007 doi: 10.1016/j.pneurobio.2007.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pietila K, Lahde T, Attila M, Ahtee L, Nordberg A. Regulation of nicotinic receptors in the brain of mice withdrawn from chronic oral nicotine treatment. Naunyn-Schmiedeberg's Arch Pharmacol. 1998;357:176–182. doi: 10.1007/pl00005152. [DOI] [PubMed] [Google Scholar]
  • 16.Collins A, Romm E, Wehner J. Dissociation of the apparent relationship between nicotine tolerance and up-regulation of nicotinic receptors. Brain Research Bulletin. 1990;25:373–379. doi: 10.1016/0361-9230(90)90222-l. [DOI] [PubMed] [Google Scholar]
  • 17.Koylu E, Demirgoren S, London E, Pogun S. Sex difference in up-regulation of nicotinic acetylcholine receptors in rat brain. Life Sci. 1997;61:PL 185–PL190. doi: 10.1016/s0024-3205(97)00665-6. [DOI] [PubMed] [Google Scholar]
  • 18.Mamede M, Ishizu K, Ueda M, et al. Temporal change in human nicotinic acetylcholine receptor after smoking cessation: 5IA SPECT study. J Nucl Med. 2007;48:1829–35. doi: 10.2967/jnumed.107.043471. [DOI] [PubMed] [Google Scholar]
  • 19.Flores C, Rogers S, Pabreza L, Wolfe B, Kellar K. A subtype of nicotinic cholinergic receptor in rat brain is composed of alpha 4 and beta 2 subunits and is up-regulated by chronic nicotine treatment. Mol Pharmacol. 1992;41:31–37. [PubMed] [Google Scholar]
  • 20.Benowitz N, Porchet H, J 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]
  • 21.Wu J, Liu Q, Yu K, et al. Roles of nicotinic acetylcholine receptor b subunits in function of human α4-containing nicotinic receptors. J Physiol. 2006;576:103–118. doi: 10.1113/jphysiol.2006.114645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Picciotto M, Zoli M, Rimondin R, et al. Acetycholine receptors containing the beta2 subunit are involved in the reinforcing properties of nicotine. Nature. 1998;391:173–177. doi: 10.1038/34413. [DOI] [PubMed] [Google Scholar]
  • 23.Walters C, Brown S, Changeux JP, Martin B, Damaj M. The β2 but not the α7 subunit of the nicotinic acetylcholine receptor is required for nicotine-conditioned place preference in mice. Psychopharmacol. 2006;184:339–344. doi: 10.1007/s00213-005-0295-x. [DOI] [PubMed] [Google Scholar]
  • 24.Shoaib M, Gommans J, Morley A, Stolerman I, Grailhe R, Changeux J. The role of nicotinic receptor beta-2 subunits in nicotine discrimination and conditioned taste aversion. Neuropharmacology. 2002;42:530–539. doi: 10.1016/s0028-3908(01)00194-0. [DOI] [PubMed] [Google Scholar]
  • 25.Epping-Jordan M, Picciotto M, Changeux J, Pich EM. Assessment of nicotinic acetylcholine receptor subunit contributions to nicotine self-administration in mutant mice. Psychopharmacology. 1999;147:25–26. doi: 10.1007/s002130051135. [DOI] [PubMed] [Google Scholar]
  • 26.Grady S, Meinerz N, Cao J, et al. Nicotinic agonists stimulate acetylcholine release from mouse interpeduncular nucleus: a function mediated by a different nAChR than dopamine release from striatum. J Neurochem. 2001;76:258–268. doi: 10.1046/j.1471-4159.2001.00019.x. [DOI] [PubMed] [Google Scholar]
  • 27.King S, Caldarone B, Picciotto M. Beta2-subunit-containing nicotinic acetylcholine receptors are critical for dopamine-dependent locomotor activation following repeated nicotine administration. Neuropharmacology. 2004;47 1:132–139. doi: 10.1016/j.neuropharm.2004.06.024. [DOI] [PubMed] [Google Scholar]
  • 28.Brunzell D, Chang J, Schneider B, Olausson P, Taylor J, Picciotto M. β2-Subunit-containing nicotinic acetylcholine receptors are involved in nicotine-induced increases in conditioned reinforcement but not progressive ratio responding for food in C57BL/6 mice. Psychopharmacology. 2006;184:328–338. doi: 10.1007/s00213-005-0099-z. [DOI] [PubMed] [Google Scholar]
  • 29.Liu X, Koren AO, Yee SK, Pechnick RN, Poland RE, London ED. Self-administration of 5-iodo-A-85380, a beta2-selective nicotinic receptor ligand, by operantly trained rats. Neuroreport. 2003;14:1503–5. doi: 10.1097/00001756-200308060-00020. [DOI] [PubMed] [Google Scholar]
  • 30.Marubio LM, del Mar Arroyo-Jimenez M, Cordero-Erausquin M, et al. Reduced antinociception in mice lacking neuronal nicotinic receptor subunits. Nature. 1999;398:805–10. doi: 10.1038/19756. [DOI] [PubMed] [Google Scholar]
  • 31.McCallum SE, Collins AC, Paylor R, Marks MJ. Deletion of the beta 2 nicotinic acetylcholine receptor subunit alters development of tolerance to nicotine and eliminates receptor upregulation. Psychopharmacology (Berl) 2006;184:314–27. doi: 10.1007/s00213-005-0076-6. [DOI] [PubMed] [Google Scholar]
  • 32.Tritto T, McCallum SE, Waddle SA, et al. Null mutant analysis of responses to nicotine: deletion of beta2 nicotinic acetylcholine receptor subunit but not alpha7 subunit reduces sensitivity to nicotine-induced locomotor depression and hypothermia. Nicotine Tob Res. 2004;6:145–58. doi: 10.1080/14622200310001656966. [DOI] [PubMed] [Google Scholar]
  • 33.Besson M, David V, Suarez S, et al. Genetic dissociation of two behaviors associated with nicotine addiction: beta-2 containing nicotinic receptors are involved in nicotine reinforcement but not in withdrawal syndrome. Psychopharmacology (Berl) 2006;187:189–99. doi: 10.1007/s00213-006-0418-z. [DOI] [PubMed] [Google Scholar]
  • 34.Sallette J, Bohler S, Benoit P, et al. An extracellular protein microdomain controls upregulation of neuronal nicotinic acetylcholine receptors by nicotine. J Biol Chem. 2004;279:18767–18775. doi: 10.1074/jbc.M308260200. [DOI] [PubMed] [Google Scholar]
  • 35.Davila-Garcia M, Musachio J, Kellar K. Chronic nicotine administration does not increase nicotinic receptors labeled by [125I]-epibatidine in adrenal gland, superior cerivical ganglia, pineal or retina. Journal of Neurochemistry. 2003;85:1237–1246. doi: 10.1046/j.1471-4159.2003.01774.x. [DOI] [PubMed] [Google Scholar]
  • 36.Flores C, Davila-Garcia M, Ulrich Y, Kellar K. Differential regulation of neuronal nicotinic receptor binding sites following chronic nicotine administration. Journal of Neurochemistry. 1997;69:2216–2219. doi: 10.1046/j.1471-4159.1997.69052216.x. [DOI] [PubMed] [Google Scholar]
  • 37.Marks M, Rowell P, Cao J, Grady S, McCallum S, Collins A. 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]
  • 38.Mao D, Perry DC, Yasuda RP, Wolfe BB, Kellar KJ. The alpha4beta2alpha5 nicotinic cholinergic receptor in rat brain is resistant to up-regulation by nicotine in vivo. J Neurochem. 2008;104:446–56. doi: 10.1111/j.1471-4159.2007.05011.x. [DOI] [PubMed] [Google Scholar]
  • 39.Mukhin A, Gundisch D, Horti A, et al. 5-Iodo-A-85380, an α4β2 subtype-selective ligand for nicotinic acetylcholine receptors. Mol Pharmacol. 2000;57:642–649. doi: 10.1124/mol.57.3.642. [DOI] [PubMed] [Google Scholar]
  • 40.Horti A, Koren A, Lee K, et al. Radiosynthesis and preliminary evaluation of 5-[123/125I]iodo-3-(2(S)-azetidinylmethoxy)pyridine: a radioligand for nicotinic acetylcholine receptors. Nucl Med Biol. 1999;26:175–182. doi: 10.1016/s0969-8051(98)00086-9. [DOI] [PubMed] [Google Scholar]
  • 41.Fujita M, Seibyl JP, Vaupel DB, et al. Whole-body biodistribution, radiation absorbed dose, and brain SPET imaging with [123i]5-i-A-85380 in healthy human subjects. Eur J Nucl Med Mol Imaging. 2002;29:183–90. doi: 10.1007/s00259-001-0695-z. [DOI] [PubMed] [Google Scholar]
  • 42.Ueda M, Iida Y, Mukai T, et al. 5-[123I]Iodo-A-85380: assessment of pharmacological safety, radiation dosimetry and SPECT imaging of brain nicotinic receptors in healthy human subjects. Annals of Nuclear Medicine. 2004;18:337–344. doi: 10.1007/BF02984473. [DOI] [PubMed] [Google Scholar]
  • 43.Staley JK, van Dyck CH, Weinzimmer D, et al. 123I-5-IA-85380 SPECT measurement of nicotinic acetylcholine receptors in human brain by the constant infusion paradigm: feasibility and reproducibility. J Nucl Med. 2005;46:1466–72. [PubMed] [Google Scholar]
  • 44.Chefer S, Horti A, Lee K, et al. In vivo imaging of brain nicotinic acetylcholine receptors with 5-[123I]iodo-A-85830 using single photon emission computed tomography. Life Sci. 1998;63:355–360. doi: 10.1016/s0024-3205(98)00514-1. [DOI] [PubMed] [Google Scholar]
  • 45.Stitzer M, Bigelow G. Contingent reinforcement for reduced breath carbon monoxide levels: target-specific effects on cigarette smoking. Addictive Behaviors. 1985;10:345–349. doi: 10.1016/0306-4603(85)90030-9. [DOI] [PubMed] [Google Scholar]
  • 46.Stitzer M, Rand C, Bigelow G, Mead A. Contingent payment procedures for smoking reduction and cessation. Journal of Applied Behavior Analysis. 1986;19:197–202. doi: 10.1901/jaba.1986.19-197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Roll J, Higgins S, Badger G. An experimental comparison of three different schedules of reinforcement of drug abstinence using cigarette smoking as an exemplar. J App Beh Analysis. 1996;29:495–504. doi: 10.1901/jaba.1996.29-495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Heatherton T, Kozlowski L, Frecker R, Fagerstrom K. The Fagerstrom test for nicotine dependence: a revision of the Fagerstrom tolerance questionairre. Brit J Addiction. 1991;86:1119–1127. doi: 10.1111/j.1360-0443.1991.tb01879.x. [DOI] [PubMed] [Google Scholar]
  • 49.Tiffany S, Drobes D. The development and initial validation of a questionnaire on smoking urges. British Journal of Addictions. 1991;86:1467–1476. doi: 10.1111/j.1360-0443.1991.tb01732.x. [DOI] [PubMed] [Google Scholar]
  • 50.Hatsukami D, Hughes J, Pickens R, Svikis D. Tobacco withdrawal symptoms: an experimental analysis. Psychopharmacology. 1984;84:231–236. doi: 10.1007/BF00427451. [DOI] [PubMed] [Google Scholar]
  • 51.Zoghbi S, Tamagnan G, Fujita M, et al. Measurement of plasma metabolites of (S)-5-[123I]iodo-3-(2-azetidinylmethoxy) pyridine (5-IA-85380), a nicotinic acetylcholine receptor imaging agent, in non-human primates. Nucl Med Biol. 2001;28:91–96. doi: 10.1016/S0969-8051(00)00188-8. [DOI] [PubMed] [Google Scholar]
  • 52.Innis RB, Cunningham VJ, Delforge J, et al. Consensus nomenclature for in vivo imaging of reversibly binding radioligands. J Cereb Blood Flow Metab. 2007;27:1533–9. doi: 10.1038/sj.jcbfm.9600493. [DOI] [PubMed] [Google Scholar]
  • 53.Malaiyandi V, Sellers EM, Tyndale RF. Implications of CYP2A6 genetic variation for smoking behaviors and nicotine dependence. Clin Pharmacol Ther. 2005;77:145–58. doi: 10.1016/j.clpt.2004.10.011. [DOI] [PubMed] [Google Scholar]
  • 54.Mwenifumbo JC, Tyndale RF. Genetic variability in CYP2A6 and the pharmacokinetics of nicotine. Pharmacogenomics. 2007;8:1385–402. doi: 10.2217/14622416.8.10.1385. [DOI] [PubMed] [Google Scholar]
  • 55.Desai HD, Seabolt J, Jann MW. Smoking in patients receiving psychotropic medications: a pharmacokinetic perspective. CNS Drugs. 2001;15:469–94. doi: 10.2165/00023210-200115060-00005. [DOI] [PubMed] [Google Scholar]
  • 56.Baldwin RM, Zoghbi SS, Staley JK, et al. Chemical fate of the nicotinic acetylcholinergic radiotracer [123I]5-IA-85380 in baboon brain and plasma. Nucl Med Biol. 2006;33:549–54. doi: 10.1016/j.nucmedbio.2006.03.002. [DOI] [PubMed] [Google Scholar]
  • 57.Lewis DF. Human cytochromes P450 associated with the phase 1 metabolism of drugs and other xenobiotics: a compilation of substrates and inhibitors of the CYP1, CYP2 and CYP3 families. Curr Med Chem. 2003;10:1955–72. doi: 10.2174/0929867033456855. [DOI] [PubMed] [Google Scholar]
  • 58.Adinoff B. Neurobiologic processes in drug reward and addiction. Harv Rev Psychiatry. 2004;12:305–20. doi: 10.1080/10673220490910844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Benwell ME, Balfour DJ. The effects of acute and repeated nicotine treatment on nucleus accumbens dopamine and locomotor activity. Br J Pharmacol. 1992;105:849–56. doi: 10.1111/j.1476-5381.1992.tb09067.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dwoskin LP, Teng L, Buxton ST, Crooks PA. (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. 1999;288:905–11. [PubMed] [Google Scholar]
  • 61.Rose JE, Behm FM, Westman EC, et al. PET studies of the influences of nicotine on neural systems in cigarette smokers. Am J Psychiatry. 2003;160:323–33. doi: 10.1176/appi.ajp.160.2.323. [DOI] [PubMed] [Google Scholar]
  • 62.Due DL, Huettel SA, Hall WG, Rubin DC. Activation in mesolimbic and visuospatial neural circuits elicited by smoking cues: evidence from functional magnetic resonance imaging. Am J Psychiatry. 2002;159:954–60. doi: 10.1176/appi.ajp.159.6.954. [DOI] [PubMed] [Google Scholar]
  • 63.McClernon FJ, Gilbert DG. Human functional neuroimaging in nicotine and tobacco research: basics, background, and beyond. Nicotine Tob Res. 2004;6:941–59. doi: 10.1080/14622200412331337394. [DOI] [PubMed] [Google Scholar]
  • 64.Smolka MN, Buhler M, Klein S, et al. Severity of nicotine dependence modulates cue-induced brain activity in regions involved in motor preparation and imagery. Psychopharmacology (Berl) 2006;184:577–88. doi: 10.1007/s00213-005-0080-x. [DOI] [PubMed] [Google Scholar]
  • 65.Brody AL, Mandelkern MA, Olmstead RE, et al. Neural substrates of resisting craving during cigarette cue exposure. Biol Psychiatry. 2007;62:642–51. doi: 10.1016/j.biopsych.2006.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Porro CA, Francescato MP, Cettolo V, et al. Primary motor and sensory cortex activation during motor performance and motor imagery: a functional magnetic resonance imaging study. J Neurosci. 1996;16:7688–98. doi: 10.1523/JNEUROSCI.16-23-07688.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cunnington R, Windischberger C, Deecke L, Moser E. The preparation and readiness for voluntary movement: a high-field event-related fMRI study of the Bereitschafts-BOLD response. Neuroimage. 2003;20:404–12. doi: 10.1016/s1053-8119(03)00291-x. [DOI] [PubMed] [Google Scholar]
  • 68.Hammond D, Fong GT, Cummings KM, Hyland A. Smoking topography, brand switching, and nicotine delivery: results from an in vivo study. Cancer Epidemiol Biomarkers Prev. 2005;14:1370–5. doi: 10.1158/1055-9965.EPI-04-0498. [DOI] [PubMed] [Google Scholar]
  • 69.Andreasen NC, O'Leary DS, Cizadlo T, et al. Remembering the past: two facets of episodic memory explored with positron emission tomography. Am J Psychiatry. 1995;152:1576–85. doi: 10.1176/ajp.152.11.1576. [DOI] [PubMed] [Google Scholar]
  • 70.Gao JH, Parsons LM, Bower JM, Xiong J, Li J, Fox PT. Cerebellum implicated in sensory acquisition and discrimination rather than motor control. Science. 1996;272:545–7. doi: 10.1126/science.272.5261.545. [DOI] [PubMed] [Google Scholar]
  • 71.Lee JH, Lim Y, Wiederhold BK, Graham SJ. A functional magnetic resonance imaging (FMRI) study of cue-induced smoking craving in virtual environments. Appl Psychophysiol Biofeedback. 2005;30:195–204. doi: 10.1007/s10484-005-6377-z. [DOI] [PubMed] [Google Scholar]
  • 72.Grant S, London ED, Newlin DB, et al. Activation of memory circuits during cue-elicited cocaine craving. Proc Natl Acad Sci U S A. 1996;93:12040–5. doi: 10.1073/pnas.93.21.12040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wang GJ, Volkow ND, Fowler JS, et al. Regional brain metabolic activation during craving elicited by recall of previous drug experiences. Life Sci. 1999;64:775–84. doi: 10.1016/s0024-3205(98)00619-5. [DOI] [PubMed] [Google Scholar]
  • 74.Schneider F, Habel U, Wagner M, et al. Subcortical correlates of craving in recently abstinent alcoholic patients. Am J Psychiatry. 2001;158:1075–83. doi: 10.1176/appi.ajp.158.7.1075. [DOI] [PubMed] [Google Scholar]
  • 75.Hester R, Garavan H. Executive dysfunction in cocaine addiction: evidence for discordant frontal, cingulate, and cerebellar activity. J Neurosci. 2004;24:11017–22. doi: 10.1523/JNEUROSCI.3321-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Jackson KJ, Martin BR, Changeux JP, Damaj MI. Differential role of nicotinic acetylcholine receptor subunits in physical and affective nicotine withdrawal signs. J Pharmacol Exp Ther. 2008;325:302–12. doi: 10.1124/jpet.107.132977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Mitsis EM, Cosgrove KP, Staley JK, et al. Age-related decline in nicotinic receptor availability with [(123)I]5-IA-85380 SPECT. Neurobiol Aging. 2008 doi: 10.1016/j.neurobiolaging.2007.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Mukhin AG, Kimes AS, Chefer SI, et al. Greater Nicotinic Acetylcholine Receptor Density in Smokers Than in Nonsmokers: A PET Study with 2-18F-FA-85380. J Nucl Med. 2008 doi: 10.2967/jnumed.108.050716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pauly JR, Marks MJ, Gross SD, Collins AC. An autoradiographic analysis of cholinergic receptors in mouse brain after chronic nicotine treatment. J Pharmacol Exp Ther. 1991;258:1127–36. [PubMed] [Google Scholar]
  • 80.Marks M, Stitzel J, Collins A. Time course study of the effects of chronic nicotine infusion on drug response and brain receptors. J Pharmacol Exp Therap. 1985;235:619–628. [PubMed] [Google Scholar]
  • 81.Ksir C, Hakan R, Hall D, Kellar K. Exposure to nicotine enhances the behavioral stimulant effect of nicotine and increases binding of [3H]-acetylcholine to nicotinic receptors. Psychopharmacology. 1985;24:527. doi: 10.1016/0028-3908(85)90058-9. [DOI] [PubMed] [Google Scholar]
  • 82.Hughes JR. Effects of abstinence from tobacco: valid symptoms and time course. Nicotine Tob Res. 2007;9:315–27. doi: 10.1080/14622200701188919. [DOI] [PubMed] [Google Scholar]
  • 83.Walters CL, Brown S, Changeux JP, Martin B, Damaj MI. The beta2 but not alpha7 subunit of the nicotinic acetylcholine receptor is required for nicotine-conditioned place preference in mice. Psychopharmacology (Berl) 2006;184:339–44. doi: 10.1007/s00213-005-0295-x. [DOI] [PubMed] [Google Scholar]
  • 84.Brunzell DH, Chang JR, Schneider B, Olausson P, Taylor JR, Picciotto MR. beta2-Subunit-containing nicotinic acetylcholine receptors are involved in nicotine-induced increases in conditioned reinforcement but not progressive ratio responding for food in C57BL/6 mice. Psychopharmacology (Berl) 2006;184:328–38. doi: 10.1007/s00213-005-0099-z. [DOI] [PubMed] [Google Scholar]
  • 85.Lai A, Parameswaran N, Khwaja M, et al. Long-term nicotine treatment decreases striatal alpha 6* nicotinic acetylcholine receptor sites and function in mice. Mol Pharmacol. 2005;67:1639–47. doi: 10.1124/mol.104.006429. [DOI] [PubMed] [Google Scholar]
  • 86.McCallum SE, Parameswaran N, Bordia T, Fan H, McIntosh JM, Quik M. Differential regulation of mesolimbic alpha 3/alpha 6 beta 2 and alpha 4 beta 2 nicotinic acetylcholine receptor sites and function after long-term oral nicotine to monkeys. J Pharmacol Exp Ther. 2006;318:381–8. doi: 10.1124/jpet.106.104414. [DOI] [PubMed] [Google Scholar]
  • 87.McCallum SE, Parameswaran N, Bordia T, et al. Increases in alpha4* but not alpha3*/alpha6* nicotinic receptor sites and function in the primate striatum following chronic oral nicotine treatment. J Neurochem. 2006;96:1028–41. doi: 10.1111/j.1471-4159.2005.03646.x. [DOI] [PubMed] [Google Scholar]
  • 88.Perez XA, Bordia T, McIntosh JM, Grady SR, Quik M. Long-term nicotine treatment differentially regulates striatal alpha6alpha4beta2* and alpha6(Nonalpha4)beta2* nAChR expression and function. Mol Pharmacol. 2008;74:844–53. doi: 10.1124/mol.108.048843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Cui C, Booker TK, Allen RS, et al. The beta3 nicotinic receptor subunit: a component of alpha-conotoxin MII-binding nicotinic acetylcholine receptors that modulate dopamine release and related behaviors. J Neurosci. 2003;23:11045–53. doi: 10.1523/JNEUROSCI.23-35-11045.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Besson M, Granon S, Mameli-Engvall M, et al. Long-term effects of chronic nicotine exposure on brain nicotinic receptors. Proc Natl Acad Sci U S A. 2007;104:8155–60. doi: 10.1073/pnas.0702698104. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES