Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Journal of Addiction Research & Therapy - Targeting the α 4 β 2- and α 7-Subtypes of Nicotinic Acetylcholine Receptors for Smoking Cessation Medication Development
ISSN: 2155-6105

Journal of Addiction Research & Therapy
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Review Article   
  • J Addict Res Ther 2019, Vol 10(2): 381
  • DOI: 10.4172/2155-6105.1000381

Targeting the α 4 β 2- and α 7-Subtypes of Nicotinic Acetylcholine Receptors for Smoking Cessation Medication Development

Lakshmi Ramachandran Nair and Xiu Liu*
Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA
*Corresponding Author: Xiu Liu, Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA, Tel: +1(601) 984-2875, Email: xliu@umc.edu

Received: 21-Jan-2019 / Accepted Date: 08-Apr-2019 / Published Date: 15-Apr-2019 DOI: 10.4172/2155-6105.1000381

Abstract

Nicotine exerts its reinforcing actions via activating the nicotinic acetylcholine receptors (nAChRs). Among an increasing number of nAChR subtypes, the α4β2 and α7 nAChRs are the two major ones, accounting for about 95% of the whole nAChR population in brain. Research findings from our own laboratory, together with other reports in the field, suggest critical and differential involvement of the α4β2 and α7 nAChRs in the process of nicotine dependence and tobacco addiction. Specifically, rat models of nicotine consumption and cue-induced relapse were used to examine the effects of selective antagonism of these two nAChR subtypes on the primary reinforcement of nicotine and the conditioned reinforcing actions of nicotine-associated environmental stimuli (cues). Results demonstrated that blockade of the α4β2 but not α7 subtype effectively reduced nicotine intake, whereas α7 but not α4β2 nAChR blockade reversed cue-triggered nicotine relapse behavior. These findings lend support for the continued effort to develop cholinergic agents aiming at the α4β2 nAChRs for reducing or stopping smoking. However, it is suggested that manipulation of α7 nAChR activity would be a promising target for preventing smoking relapse triggered by exposure to environmental cues.

Keywords: Conditioned stimuli (cues); Nicotinic acetylcholine receptors (nachrs); Reinforcing actions; Relapse; Self-administration

Introduction

Tobacco-related diseases are a major problem in many perspectives from human health to social economics [1]. For example, in the United States tobacco smoking becomes a leading preventable cause of premature death. Every year, tobacco smoking results in the loss of 450,000 lives and economic cost of $289 to $333 billion [2]. Currently, approximately 42.1 million American adults are smokers, representing about 18.1% of the population [3]. Although almost all smokers want to quit smoking and make attempts, up to 97% of them relapse to tobacco smoking [3-6]. The high relapse rates of tobacco smoking present a formidable challenge for the success of smoking cessation efforts including currently available pharmacotherapies (e.g., nicotine replacements, bupropion, and varenicline).

Nicotinic acetylcholine receptors (nAChRs) mediate the pharmacological (including its reinforcing) actions of nicotine. These receptors are ion channels composed of five subunits. There are twelve nAChR subunits: nine α-subunits (α2-α10) and three β-subunits (β2- β4). These subunits assemble the nAChRs into either heteromeric (α- and β-subunits) or homomeric (α-subunit only) combinations [7-10]. Increasing number of subtypes of the nAChRs has been and will be identified, among which the heteromeric α4β2- and homomeric α7- containing receptors are the most abundant and widespread subtypes, comprising about 95% of total nAChRs in the brain [7,8,11-14]. These two subtypes show differences in their localization, density, and functional characteristics (e.g., kinetics of activation, desensitization, and recovery from desensitization, and Ca2+ permeability) [9,15-18]. Many studies including our own research have demonstrated a critical role of the α4β2 nAChRs in mediating the primary reinforcing actions of nicotine [19-23], while, in contrast, a lack of a clear role of the α7 nAChRs in the nicotine reinforcement [24, 25-31].

Increasing clinical observations and laboratory animal studies have demonstrated the conditioned incentive properties of drug-associated environmental stimuli (cues) [32-37]. In smokers, the environmental cues related to smoking behavior including both distally situational contexts and proximal sensory cues such as the visual and olfactory stimuli associated with each puff elicit subjective states that can trigger smoking and nicotine-seeking behavior [32,33,38-49]. In animal research, the ability of nicotine-related cues to reinstate nicotineseeking behavior has been well documented [31,37,50-59]. A great interest has focused on the investigation of neurobiological mechanisms underlying the conditioned motivational effects of nicotine cues [36 for a recent review].

Our animal research work over the past decade or so has demonstrated the role of nicotinic neurotransmission in the mediation of the conditioned motivational effects of nicotine-associated cues. The method used for testing the behavior motivational effects of nicotine cues and other relapse risk factors (e.g., stress or drug priming) was the response-reinstatement tests, which has been validated to be an animal model of relapse [60-62]. Using this testing procedure, we for the first time found that a nonselective nAChR antagonist mecamylamine effectively reversed the cue-induced reinstatement of nicotine-seeking behavior [63]. And furthermore, our recent work has demonstrated that the α7 but not α4β2 nAChRs mediate the cue-induced reinstatement of nicotine-seeking behavior [31].

Experimental Procedures for Testing Nicotine Consumption and Relapse

Rats were used for testing nicotine intake and relapse behavior. After implantation of an indwelling intravenous catheter, the animals were trained to self-administer nicotine in daily one-hour sessions in the standard operant conditioning chambers. In the sessions, once the rats reached a fixed-ratio 5 schedule requirement of responses on the active lever, an infusion of nicotine was delivered. To establish a nicotine-conditioned cue, each nicotine infusion was paired with presentation of a sensory stimulus. For testing cue-triggered relapse behavior, extinction sessions were performed after completion of the self-administration and conditioning training. In these sessions, responses on the lever produced no programed outcomes. After responding was extinguished, the response-reinstatement test sessions were conducted where lever-pressing responses led to the cue presentations while without the delivery of nicotine [54-55,63]. The antagonists of the nAChRs were administered to the rats prior to the self-administration and the reinstatement test sessions [31,63]. The antagonists included a nonselective antagonist mecamylamine, a α4β2- selective antagonist dihydro-β-erythroidine (DhβE), and a α7-selective antagonist methyllycaconitine (MLA).

Distinct Roles of the Α4β2 and Α7 Nachrs in the Reinforcement by Nicotine versus Conditioned Motivation by Nicotine Cues

Ample evidence obtained from both human and animal studies has demonstrated a clear role of the α4β2 nAChRs in mediating the primary reinforcement by nicotine [19-23]. In contrast, however, it is not quite clear whether the α7 nAChRs play a role in nicotine primary reinforcement [24,25-31]. For example, in one study MLA did not interfere with nicotine self-administration [24], whereas, in another report MLA produced a suppressant effect [64]. Conditioned place preference studies also excluded a possible role for α7 nAChRs in the mediation of nicotine reward. For example, mice that were either treated with MLA or deficient in α7 nAChRs developed nicotineinduced conditioned place preference at a level similar to their control counterparts [28,65]. Interestingly, Brunzell and McIntosh [29] found that the α7 nAChR-selective antagonist α-conotoxin ArlB [VIIL, VI6D], when microinjected into rat nucleus accumbens shell and anterior cingulate cortex, significantly increased nicotine selfadministration behavior under a progressive-ratio schedule of reinforcement. Of significance is our recent research showing that MLA did not change the self-administration of nicotine [31], indicating that activation of the α7 nAChRs is not required for the reinforcement by nicotine. In summary, activation of α7 nAChRs is proposed to play little, if any at all, role in the mediation of nicotine primary reinforcement.

In the response-reinstatement tests, response-contingent presentation of the nicotine-conditioned cues triggered the recovery of lever-press responding after extinction. Such an effect was specific for nicotine-seeking behavior in that responses on the inactive lever remained unchanged, indicating the unlikelihood of a result of nonspecific behavioral arousal. The conditioned incentive properties of nicotine cues have been very well documented in literature including our own series of studies over the last decade or so [31,37,50-59]. These results obtained from animal research lend support for clinical observations that smoking-related cues enhance desire to smoke [32,33,38-49]. Together, these findings suggest that re-exposure to environmental stimuli previously associated with nicotine intake can play an important role in relapse to tobacco smoking in abstinent smokers.

The nonselective nAChR antagonist mecamylamine effectively reversed the recovery of nicotine-seeking behavior triggered by nicotine cue presentation. This finding demonstrates the requirement of neurotransmission via the nAChRs for the expression of cuetriggered relapse to smoking behavior. It is consistent with clinical observations. For example, mecamylamine was reported to decrease the likeness for intravenously infused nicotine in smokers [66], craving for smoking [67], and satisfaction following smoking [68-70].

In contrast to the role of the α4β2 nAChRs in mediating nicotine reinforcement, our studies showed that blockade of these receptors by DHβE pre-treatment did not interfere with the cue-induced reinstatement of nicotine-seeking responses [31]. The doses used should be sufficient to antagonize the receptors because such a dose range has often been used in the literature, including selfadministration studies [24,71,72] and our own previous study showing its suppressant effect on nicotine-enhanced lever-pressing behavior in response to the presentation of a reinforcing stimulus [73]. These results are consistent with other studies. For example, varenicline, a partial agonist at α4β2 nAChRs, had no effect on the cue-induced reinstatement of nicotine seeking assessed using similar extinctionreinstatement procedures in rodents [74,75] and did not change cuespecific craving in smokers [76]. However, blockade of the α4β2 nAChRs by (DhβE did not change the cue-triggered recovery of nicotine-seeking behavior. That indicates the lack of a role of the neurotransmission via the α4β2 nAChRs in mediating the conditioned incentive motivation by nicotine cue exposure.

It is interesting to note that blockade of the α7 nAChRs by MLA dose-dependently reduced the cue-triggered recovery of nicotineseeking behavior. It demonstrates the requirement of the activation of the α7 nAChRs mediating the conditioned incentive motivation by exposure to nicotine-conditioned environmental cues. Since neither did MLA alter the recovery of cue-triggered food-seeking behavior nor changed the enhancing effect of nicotine on other intrinsically reinforcing sensory stimulus [73], MLA acted specifically at the nicotine cue without interference with general locomotor activity, arousal state, the motivation to earn rewards, and operant goaldirected behavior., Therefore, the specific inhibitory effect of MLA on the cue-induced resumption of nicotine seeking suggests that activation of α7 nAChRs is necessary for the expression of conditioned incentive motivation induced by nicotine-related cues. Our recent studies (not yet published) further demonstrated that α-conotoxin ArlB [VIIL, VI6D] microinjected into the nucleus accumbens but not ventral tegmental area effectively blocked the cue-triggered reinstatement of nicotine-seeking responses. The finding indicates that the nucleus accumbens is a critical neuroanatomical substrate for the α7 nAChRs to mediate the behavioral effect of nicotine cues.

Implications for Development of Smoking Cessation Medications

Converging experimental evidence suggests that the α4β2 and α7 nAChRs play differential roles in mediating the reinforcing actions of nicotine versus the conditioned incentive properties of nicotine cues. The α4β2 nAChRs participate in nicotine primary reinforcement but not conditioned reinforcement induced by nicotine cues, whereas α7 nAChRs do the opposite. The differential involvement of these two nAChR subtypes indicates a dissociation of the neurobiological mechanisms that underlie the primary reinforcing actions of nicotine and secondary reinforcement induced by nicotine cues. High level of α7 nAChRs is expressed in the cortico mesolimbic circuits, including profrontal cortex, ventral tegmental area and nucleus accumbens as well as the hippocampus and hypothalamus [18,77-79]. These receptors may play an essential role in the modulation of dopamine rewarding pathways in that agonists produce cognition enhancement [80]. That may underlie the role of α7 nAChRs in mediating the conditioned incentive properties of nicotine cues. Interestingly, similar dissociations were also demonstrated with other drugs of abuse. For example, pharmacological antagonism of opioid receptors decreased cue-induced resumption of nicotine seeking but did not alter nicotine intake [81]. Inhibition of nitric oxide synthesis reduced cue-triggered recovery of alcohol seeking but not the consumption of alcohol [82]. Blockade of orphan sigma-1 receptors decreased cue-induced recovery of cocaine-seeking responses produced no effect on cocaine intake [83]. In summary, the reinforcement of nicotine and the conditioned motivation by nicotine cues involve distinct neurobiological mechanisms.

The research work reviewed above supports the continued effort to develop nicotinic agents aiming at the α4β2 nAChRs for reducing and eventually stopping nicotine consumption and tobacco smoking. The α4β2 targeted medications have found increasing clinical use although their efficacy is not yet quite satisfactory [84-86]. However, the lack of involvement of the α4β2 nAChRs in nicotine cue effect might help explain the inability of currently available smoking cessation medications (nicotine replacement, bupropion, and varenicline) to suppress cue-reactivity in abstinent subjects [74,75,87-90]. because these medications are full (nicotine) or partial (varenicline) agonist or antagonist (one of bupropion’s actions) at the α4β2 nAChRs. In light of the fact that cholinergic neurotransmission via the α7 subtype of nAChRs plays a critical role in mediating the conditioned incentive properties of nicotine cues, it is suggested that developing cholinergic agents aiming at the α7 nAChRs may prove to be a good strategy to prevent smoking relapse triggered by exposure to environmental cues.

These preclinical research results would have significant implications for developing medication strategies to prevent relapse in abstinent smokers. To our knowledge, however, there has been no clinical trial performed to test the potential of α7 nAChR antagonists for smoking relapse prevention. Although there have been many studies to examine characteristics of the brain nAChRs [7-18], it is lack of direct comparison of these receptors such as the α4β2 and α7 subtypes among different species, e.g., rodents versus humans. These facts call for more research effort to address the issues and acknowledge the caveats for clinical tests, if any in the future.

Acknowledgements

The research work reviewed in this chapter was supported by NIH grants R01 DA017288 and R01 DA037277 from the National Institute on Drug Abuse as well as the State of California Tobacco-Related Disease Research Program grant #12KT-0188. The authors would like to thank Courtney Jernigan, Lisa Biswas, Erin Harrison, Ramachandram Avusula, Thomas Rousselle, Thuy Tran, and Brooke Hobbs for their excellent technical assistance.

References

  1. Holford TR, Meza R, Warner KE, Meernik C, Jeon J, et al. (2014) Tobacco control and the reduction in smoking-related premature deaths in the United States, 1964-2012. JAMA 311: 164-171.
  2. Jamal A, Phillips E, Gentzke AS, Homa DM1, Babb SD, et al. (2014) Current cigarette smoking among adults-United States. Morb. Mortal. Wkly Rep. 63: 29-34.
  3. Shiffman S, Mason KM, Henningfield JE (1998) Tobacco dependence treatments: review and prospectus. Annu Rev Public Health 19: 335-358.
  4. Hughes JR, Keely J, Naud S (2004) Shape of the relapse curve and long-term abstinence among untreated smokers. Addiction 99: 29-38
  5. Gotti C, Clementi F (2004) Neuronal nicotinic receptors: from structure to pathology. Prog Neurobiol 74: 363-396.
  6. Sargent PB (1993) The diversity of neuronal nicotinic acetylcholine receptors. Annu Rev Neurosci 16: 403-443.
  7. McGehee DS, Role LW (1995) Physiological diversity of nicotinic acetylcholine receptors expressed by vertebrate neurons. Annu Rev Physiol 57: 521-546.
  8. Dani JA, Bertrand D (2007) Nicotinic acetylcholine receptors and nicotinic cholinergic mechanisms of the central nervous system. Annu Rev Pharmacol Toxicol 47: 699-729.
  9. Zoli M, Léna C, Picciotto MR, Changeux JP (1998) Identification of four classes of brain nicotinic receptors using beta2 mutant mice. J Neurosci 18: 4461-4472.
  10. Flores CM, Rogers SW, Pabreza LA, Wolfe BB, Kellar KJ (1992) 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 41: 31-37
  11. Albuquerque EX, Pereira EF, Alkondon M, Rogers SW (2009) Mammalian nicotinic acetylcholine receptors: from structure to function. Physiological Rev 89: 73-120.
  12. Millar NS, Gotti C (2009) Diversity of vertebrate nicotinic acetylcholine receptors. Neuropharmacology 56: 237-246.
  13. Albuquerque EX, Alkondon M, Pereira EF, Castro NG, Schrattenholz A, et al. (1997) Properties of neuronal nicotinic acetylcholine receptors: pharmacological characterization and modulation of synaptic function. J Pharmacol Exp Ther 280: 1117-1136.
  14. Alkondon M, Albuquerque EX (2001) Nicotinic acetylcholine receptor alpha7 and alpha4beta2 subtypes differentially control GABAergic input to CA1 neurons in rat hippocampus. J Neurophysiol 86: 3043-3055.
  15. Tribollet E, Bertrand D, Raggenbass M (2001) Role of neuronal nicotinic receptors in the transmission and processing of information in neurons of the central nervous system. Pharmacol Biochem Behav 70: 457-466.
  16. Séguéla P, Wadiche J, Dineley-Miller K, Dani JA, Patrick JW (1993) Molecular cloning, functional properties, and distribution of rat brain alpha 7: a nicotinic cation channel highly permeable to calcium. J Neurosci 13: 596-604.
  17. Vieyra-Reyes P, Picciotto MR, Mineur YS (2008) Voluntary oral nicotine intake in mice down-regulates GluR2 but does not modulate depression-like behaviors. Neurosci Lett 434: 18-22.
  18. Wonnacott S, Sidhpura N, Balfour DJ (2005) Nicotine: from molecular mechanisms to behaviour. Curr Opin Pharmacol 5: 53-59.
  19. Tapper AR, McKinney SL, Nashmi R, Schwarz J, Deshpande P, et al. (2004) Nicotine activation of alpha4* receptors: sufficient for reward, tolerance, and sensitization. Science 306: 1029-1032.
  20. Exley R, Cragg SJ (2008) Presynaptic nicotinic receptors: a dynamic and diverse cholinergic filter of striatal dopamine neurotransmission. Br J Pharmacol 153: S283-297.
  21. Mineur, YS, Picciotto MR (2008) Genetics of nicotinic acetylcholine receptors: Relevance to nicotine addiction. Biochem Pharmacol 75: 323-333.
  22. Grottick AJ, Trube G, Corrigall WA, Huwyler J, Malherbe P, et al. (2000) Evidence that nicotinic alpha (7) receptors are not involved in the hyperlocomotor and rewarding effects of nicotine. J Pharmacol Exp Ther 294: 1112-1119.
  23. Stolerman IP, Chamberlain S, Bizarro L, Fernandes C, Schalkwyk L, et al. (2004) The role of nicotinic receptor alpha 7 subunits in nicotine discrimination. Neuropharmacology 46: 363-371.
  24. Van Haaren F, Anderson KG, Haworth SC, Kem WR (1999) GTS-21, a mixed nicotinic receptor agonist/antagonist, does not affect the nicotine cue. Pharmacol Biochem Behav 64: 439-444.
  25. Pons S, Fattore L, Cossu G, Tolu S, Porcu E (2008) Crucial role of alpha4 and alpha6 nicotinic acetylcholine receptor subunits from ventral tegmental area in systemic nicotine self-administration. J Neurosci 28: 12318-12327.
  26. Walters CL, Brown S, Changeux JP, Martin B, Damaj MI (2006) The beta2 but not alpha7 subunit of the nicotinic acetylcholine receptor is required for nicotine-conditioned place preference in mice. Psychopharmacology (Berl) 184: 339-44.
  27. Brunzell DH, McIntosh JM (2012) Alpha7 nicotinic acetylcholine receptors modulate motivation to self-administer nicotine: implications for smoking and schizophrenia. Neuropsychopharmacology 37: 1134-1143.
  28. Besson M, David V, Baudonnat M, Cazala P, Guilloux JP, et al. (2012) Alpha7-nicotinic receptors modulate nicotine-induced reinforcement and extracellular dopamine outflow in the mesolimbic system in mice. Psychopharmacology (Berl) 220: 1-14.
  29. Liu X (2014) Effects of blockade of alpha4beta2 and alpha7 nicotinic acetylcholine receptors on cue-induced reinstatement of nicotine-seeking behaviour in rats. Int J Neuropsychopharmacol 17: 105-116.
  30. O'Brien CP, Childress AR, Ehrman R, Robbins SJ (1998) Conditioning factors in drug abuse: Can they explain compulsion? J Psychopharmacol. 12: 15-22.
  31. Caggiula AR, Donny EC, White AR, Chaudhri N, Booth S, et al. (2001) Cue dependency of nicotine self-administration and smoking. Pharmacol Biochem Behav 70: 515-530.
  32. Conklin, CA, Kenneth AP, Nathalie R, Joseph MCF, Ronald PS (2010) Bringing the real world into the laboratory: personal smoking and nonsmoking environments. Drug Alcohol Depend 111: 58-63.
  33. Perry CJ, Zbukvic I, Kim JH, Lawrence AJ (2014) Role of cues and contexts on drug-seeking behaviour. Br J Pharmacol 171: 4636-4672.
  34. Stoker AK, Markou A (2015) Neurobiological Bases of Cue- and Nicotine-induced Reinstatement of Nicotine Seeking: Implications for the Development of Smoking Cessation Medications. Curr Top Behav Neurosci 24: 125-154.
  35. Liu X (2010) Contribution of drug cue, priming, and stress to reinstatement of nicotine-seeking behavior in a rat model of relapse. 101: 143-163.
  36. Rose JE (2006) Nicotine and nonnicotine factors in cigarette addiction. Psychopharmacology (Berl) 184: 274-285.
  37. Niaura RS, Rohsenow DJ, Binkoff JA, Monti PM, Pedraza M, et al. (1988) Relevance of cue reactivity to understanding alcohol and smoking relapse. J Abnorm Psychol 97: 133-152.
  38. Carter BL, Tiffany ST (1999) Meta-analysis of cue-reactivity in addiction research. Addiction 94: 327-340.
  39. Miranda R Jr, Rohsenow DJ, Monti PM, Tidey J, Ray L (2008) Effects of repeated days of smoking cue exposure on urge to smoke and physiological reactivity. Addict Behav 33: 347-353.
  40. Tong C, Bovbjerg DH, Erblich J (2007) Smoking-related videos for use in cue-induced craving paradigms. Addict Behav 32: 3034-3044.
  41. Conklin CA, Robin N, Perkins KA, Salkeld RP, McClernon FJ (2008) Proximal versus distal cues to smoke: the effects of environments on smokers' cue-reactivity. Exp Clin Psychopharmacol 16: 207-214.
  42. Zhou X, Nonnemaker J, Sherrill B, Gilsenan AW, Coste F, et al. (2009) Attempts to quit smoking and relapse: factors associated with success or failure from the ATTEMPT cohort study. Addict Behav 34: 365-373.
  43. Parker AB, Gilbert DG (2008) Brain activity during anticipation of smoking-related and emotionally positive pictures in smokers and nonsmokers: a new measure of cue reactivity. Nicotine Tob Res 10: 1627-1631.
  44. Conklin CA, Vella EJ, Joyce CJ, Salkeld RP, Perkins KA, et al. (2015) Examining the relationship between cue-induced craving and actual smoking. Exp Clin Psychopharmacol 23: 90-96
  45. Carpenter MJ, Saladin ME, Larowe SD, McClure EA, Simonian S, et al. (2014) Craving, cue reactivity, and stimulus control among early-stage young smokers: effects of smoking intensity and gender. Nicotine Tob Res 16: 208-215.
  46. Veilleux JC, Skinner KD (2015) Smoking, food, and alcohol cues on subsequent behavior: a qualitative systematic review. Clin Psychol Rev 36: 13-27.
  47. Rees VW, Kreslake JM, Wayne GF, O'Connor RJ, Cummings KM, et al. (2012) Role of cigarette sensory cues in modifying puffing topography. Drug Alcohol Depend 124: 1-10.
  48. LeSage MG, Burroughs D, Dufek M, Keyler DE, Pentel PR (2004) Reinstatement of nicotine self-administration in rats by presentation of nicotine-paired stimuli, but not nicotine priming. Pharmacol Biochem Behav 79: 507-513.
  49. Cohen C, Perrault G, Griebel G, Soubrié P (2005) Nicotine-associated cues maintain nicotine-seeking behavior in rats several weeks after nicotine withdrawal: reversal by the cannabinoid (CB1) receptor antagonist, rimonabant (SR141716). Neuropsychopharmacology 30: 145-155.
  50. Paterson NE, Froestl W, Markou A (2005) Repeated administration of the GABAB receptor agonist CGP44532 decreased nicotine self-administration, and acute administration decreased cue-induced reinstatement of nicotine-seeking in rats. Neuropsychopharmacology 30: 119-128.
  51. Chiamulera C1, Tedesco V, Zangrandi L, Giuliano C, Fumagalli G (2010) Propranolol transiently inhibits reinstatement of nicotine-seeking behaviour in rats. J Psychopharmacol 24: 389-395.
  52. Liu X, Caggiula AR, Palmatier MI, Donny EC, Sved AF (2008) Cue-induced reinstatement of nicotine-seeking behavior in rats: effect of bupropion, persistence over repeated tests, and its dependence on training dose. Psychopharmacology (Berl) 196: 365-375.
  53. Liu X, Caggiula AR, Yee SK, Nobuta H, Poland RE, et al. (2006) Reinstatement of nicotine-seeking behavior by drug-associated stimuli after extinction in rats. Psychopharmacology (Berl) 184: 417-425.
  54. Abdolahi A, Acosta G, Breslin FJ, Hemby SE, Lynch WJ (2010) Incubation of nicotine seeking is associated with enhanced protein kinase A-regulated signaling of dopamine- and cAMP-regulated phosphoprotein of 32 kDa in the insular cortex. Eur J Neurosci 31: 733-741.
  55. Feltenstein MW, Ghee SM, See RE (2012) Nicotine self-administration and reinstatement of nicotine-seeking in male and female rats. Drug Alcohol Depend 121: 240-246.
  56. Fowler CD, Kenny PJ (2011) Intravenous nicotine self-administration and cue-induced reinstatement in mice: effects of nicotine dose, rate of drug infusion and prior instrumental training. Neuropharmacology 61: 687-698.
  57. Trigo JM, Le Foll B (2016) Inhibition of monoacylglycerol lipase (MAGL) enhances cue-induced reinstatement of nicotine-seeking behavior in mice. Psychopharmacology (Berl). 233: 1815-1822.
  58. Shaham Y, Shalev U, Lu L, de Wit H, Stewart J (2003) The reinstatement model of drug relapse: history, methodology and major findings. Psychopharmacology (Berl) 168: 3-20.
  59. Bossert JM, Marchant NJ, Calu DJ, Shaham Y (2013) The reinstatement model of drug relapse: recent neurobiological findings, emerging research topics, and translational research. Psychopharmacology 229: 453-476.
  60. Epstein DH, Preston KL, Stewart J, Shaham Y (2006) toward a model of drug relapse: an assessment of the validity of the reinstatement procedure. Psychopharmacology 189: 1-16.
  61. Liu X, Caggiula AR, Yee SK, Nobuta H, Sved AF, et al. (2007) Mecamylamine attenuates cue-induced reinstatement of nicotine-seeking behavior in rats. Neuropsychopharmacology 32: 710-718.
  62. Markou A, Paterson NE (2001) The nicotinic antagonist methyllycaconitine has differential effects on nicotine self-administration and nicotine withdrawal in the rat. Nicotine Tob Res 3: 361-373.
  63. Grabus SD, Martin BR, Brown SE, Damaj MI (2006) Nicotine place preference in the mouse: influences of prior handling, dose and strain and attenuation by nicotinic receptor antagonists. Psychopharmacology 184: 456-463.
  64. Rose Je, Levin Ed, Behm Fm, Westman Ec, Stein Rm, Et Al. (1995) Combined administration of agonist-antagonist as a method of regulating receptor activation. Ann N Y Acad Sci 757: 218-221
  65. Rose JE, Sampson A, Levin ED, Henningfield JE (1989) Mecamylamine increases nicotine preference and attenuates nicotine discrimination. Pharmacol Biochem Behav 32: 933-938.
  66. Rose JE, Behm FM, Westman EC, Levin ED, Stein RM et al. (1994) Mecamylamine combined with nicotine skin patch facilitates smoking cessation beyond nicotine patch treatment alone. Clin Pharmacol Ther 56: 86-99.
  67. Nemeth-Coslett R, Henningfield J, O'Keeffe M, Griffiths RR (1986) Effects of mecamylamine on human cigarette smoking and subjective ratings. Psychopharmacology 88: 420-425.
  68. Lundahl LH, Henningfield JE, Lukas SE (2000) Mecamylamine blockade of both positive and negative effects of IV nicotine in human volunteers. Pharmacol Biochem Behav 66: 637-643.
  69. Watkins SS, Epping-Jordan MP, Koob GF, Markou A (1999) Blockade of nicotine self-administration with nicotinic antagonists in rats. Pharmacol Biochem Behav 62: 743-751.
  70. Paterson NE, Min W, Hackett A, Lowe D, Hanania T, et al. (2010) The high-affinity nAChR partial agonists varenicline and sazetidine-A exhibit reinforcing properties in rats. Prog Neuropsychopharmacol Biol Psychiatry 34: 1455-1464.
  71. Liu X, Palmatier MI, Caggiula AR, Donny EC, Sved AF (2007) Reinforcement enhancing effect of nicotine and its attenuation by nicotinic antagonists in rats. Psychopharmacology 194: 463-473.
  72. O’Connor EC, Parker D, Rollema H, Mead AN (2010) The alpha4beta2 nicotinic acetylcholine-receptor partial agonist varenicline inhibits both nicotine self-administration following repeated dosing and reinstatement of nicotine seeking in rats. Psychopharmacology 208: 365-376.
  73. Wouda JA, Riga D, De Vries W, Stegeman M, van Mourik Y et al. (2011) Varenicline attenuates cue-induced relapse to alcohol, but not nicotine seeking, while reducing inhibitory response control. Psychopharmacology 216: 267-277.
  74. Gass JC, Wray JM, Hawk LW, Mahoney MC, Tiffany ST (2012) Impact of varenicline on cue-specific craving assessed in the natural environment among treatment-seeking smokers. Psychopharmacology 223: 107-116.
  75. Mansvelder HD, De Rover M, McGehee DS, Brussaard AB (2003) Cholinergic modulation of dopaminergic reward areas: upstream and downstream targets of nicotine addiction. Eur J Pharmacol 480: 117-123.
  76. Melis M, Scheggi S, Carta G, Madeddu C, Lecca S, et al. (2013) PPARalpha regulates cholinergic-driven activity of midbrain dopamine neurons via a novel mechanism involving alpha7 nicotinic acetylcholine receptors. J Neurosci 33: 6203-6211.
  77. del Toro ED, Juiz JM, Peng X, Lindstrom J, Criado M (1994) Immunocytochemical localization of the alpha 7 subunit of the nicotinic acetylcholine receptor in the rat central nervous system. J Comp Neurol 349: 325-342.
  78. Briggs CA, Grønlien JH, Curzon P, Timmermann DB, Ween H (2009) Role of channel activation in cognitive enhancement mediated by alpha7 nicotinic acetylcholine receptors. Br J Pharmacol 158: 1486-1494.
  79. Liu X, Palmatier MI, Caggiula AR, Sved AF, Donny EC, et al. (2009) Naltrexone attenuation of conditioned but not primary reinforcement of nicotine in rats. Psychopharmacology 202: 589-598.
  80. Liu X, Weiss F (2004) Nitric oxide synthesis inhibition attenuates conditioned reinstatement of ethanol-seeking, but not the primary reinforcing effects of ethanol. Alcohol Clin Exp Res 28: 1194-1199.
  81. Martin-Fardon R, Maurice T, Aujla H, Bowen WD, Weiss F (2007) Differential effects of sigma1 receptor blockade on self-administration and conditioned reinstatement motivated by cocaine vs natural reward. Neuropsychopharmacology 32: 1967-1973.
  82. Cahill K, Stevens S, Perera R, Lancaster T (2013) Pharmacological interventions for smoking cessation: an overview and network meta-analysis. Cochrane Database Syst Rev 5: CD009329.
  83. Dwoskin LP, Smith AM, Wooters TE, Zhang Z, Crooks PA, et al. (2009) Nicotinic receptor-based therapeutics and candidates for smoking cessation. Biochem Pharmacol 78: 732-743.
  84. Aubin HJ, Karila L, Reynaud M (2011) Pharmacotherapy for smoking cessation: present and future. Curr Pharm Des 17 (14), 1343-50.
  85. Hussain S, Zawertailo L, Busto U, Zack M, Farvolden P, et al. (2010) The impact of chronic bupropion on plasma cotinine and on the subjective effects of ad lib smoking: a randomized controlled trial in unmotivated smokers. Addict Behav 35: 164-167.
  86. Morissette SB, Palfai TP, Gulliver SB, Spiegel DA, Barlow DH (2005) Effects of transdermal nicotine during imaginal exposure to anxiety and smoking cues in college smokers. Psychol Addict Behav 19: 192-198.
  87. Tiffany ST, Cox LS, Elash CA (2000) Effects of transdermal nicotine patches on abstinence-induced and cue-elicited craving in cigarette smokers. J Consult Clin Psychol 68: 233-240.
  88. Waters AJ, Shiffman S, Sayette MA, Paty JA, Gwaltney CJ, et al. (2004) Cue-provoked craving and nicotine replacement therapy in smoking cessation. J Consult Clin Psychol 72: 1136-1143.

Citation: Nair LR, Liu X (2019) Targeting the α4β2- and α7-Subtypes of Nicotinic Acetylcholine Receptors for Smoking Cessation Medication Development. J Addict Res Ther 10:381. DOI: 10.4172/2155-6105.1000381

Copyright: © 2019 Nair LR, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.

Top