Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 20.
Published in final edited form as: CNS Neurol Disord Drug Targets. 2011 Sep 1;10(6):651–658. doi: 10.2174/187152711797247849

Targeting nicotinic receptors for Parkinson's disease therapy

Maryka Quik 1,*, Tanuja Bordia 1, Luping Huang 1, Xiomara Perez 1
PMCID: PMC3748273  NIHMSID: NIHMS503961  PMID: 21838678

Abstract

A promising target for improved therapeutics in Parkinson's disease is the nicotinic acetylcholine receptor (nAChR). nAChRs are widely distributed throughout the brain, including the nigrostriatal system, and exert important modulatory effects on numerous behaviors. Accumulating evidence suggests that drugs such as nicotine that act at these sites may be of benefit for Parkinson's disease treatment. Recent work indicates that a potential novel therapeutic application is the use of nicotine to reduce levodopa-induced dyskinesias, a side effect of dopamine replacement therapy for Parkinson's disease. Several clinical trials also report that nicotine may diminish disease symptoms. Not only may nAChR drugs provide symptomatic improvement, but they may also attenuate the neurodegenerative process itself. This latter idea is supported by epidemiological studies which consistently demonstrate a ~50% reduced incidence of Parkinson's disease in smokers. Experimental work in parkinsonian animal models suggests that nicotine in tobacco may contribute to this protection. These combined findings suggest that nicotine and nAChR drugs offer the possibility of improved therapeutics for Parkinson's disease.

Keywords: Nicotine, nicotinic receptors, levodopa, dyskinesias, neuroprotection, parkinsonian, Parkinson's disease

INTRODUCTION

Parkinson's disease is a neurodegenerative movement disorder characterized by a generalized neuronal decline in the CNS, with a particularly prominent loss of nigrostriatal dopaminergic neurons 1. Dopamine replacement provides excellent therapeutic relief in the early stages of the disease. However, there is a constant search for new pharmacotherapies for Parkinson's disease management because of limitations associated with current treatment 1-2. Effectiveness in reducing Parkinson's disease symptoms diminishes with continued use and side effects develop, including on-off phenomena and dyskinesias. Another drawback with current treatment is that it primarily provides symptomatic relief, with an inevitable disease progression.

Emerging studies suggest that nicotine treatment might be a useful adjunct therapy for Parkinson's disease. Recent work in monkey, mouse and rat parkinsonian animal models show that nicotine reduces levodopa-induced dyskinesias, a side effect of dopamine replacement therapy that may be as debilitating as the disease itself 3. Clinical studies also suggest that nicotine has antiparkinsonian effects 4. Lastly, nicotine protects against nigrostriatal damage in different experimental models 3a, 5. These latter findings form the basis for the suggestion that nicotine in tobacco products may be associated with the decreased incidence of Parkinson's disease observed with smoking 4, 6.

The goal of this paper is to discuss the interaction between the dopaminergic and nicotinic cholinergic systems to provide a framework for understanding a role for nicotine in Parkinson's disease. We also review some promising pre-clinical work which indicates that nicotine and/or nAChR drugs may be useful for improving levodopa-induced dyskinesias in Parkinson's disease and for protection against long-term nigrostriatal damage.

RELATIONSHIP BETWEEN THE NICOTINIC CHOLINERGIC AND DOPAMINERIGIC SYSTEM

Knowledge of the anatomical interrelationship between the striatal cholinergic and dopaminergic systems has contributed significantly to our understanding of the role of nicotine in Parkinson's disease therapy. Numerous studies have shown that the terminals from the ascending nigrostriatal dopaminergic projections from the substantia nigra extensively overlap with acetylcholine interneurons present in the striatum 7. These cholinergic neurons represent a small percentage (~2-3%) of the neuronal cell bodies in the striatum, the greater majority (~95%) of which are GABAergic. However, despite their limited number, the cholinergic neurons are quite large. Importantly, their dendritic arborization forms a dense network in striatum whose distribution closely matches that of the dopaminergic terminals, with a very similar expression pattern of dopaminergic (dopamine, tyrosine hydroxylase, dopamine transporter) and cholinergic (acetylcholine, choline acetyltransferase and acetylcholinesterase) markers 8. Cholinergic interneurons tonically secrete acetylcholine, which modulates neurotransmitter release from striatal dopaminergic neurons, and also to a lesser extent from cortical glutamatergic terminals. Acetylcholine exerts these effects on dopamine release via an interaction at nAChRs, of which there are multiple subtypes 9.

NICOTINIC RECEPTOR SUBTYPES

Structurally the CNS nAChR has as its basic motif, a pentamer of five subunits around a central pore that lies within the membrane bilayer. Molecular cloning has identified nine α (α2-α10) and three β (β2-β4) subunits. Select α (α2-α6) and β (β2-β4) subunits assemble into heteropentamers. Homopentamers composed only of α subunits also exist and may contain only α7, α8 and α9 subunits or combinations of α7α8 and α9α10 subunits 9. The binding of acetylcholine to the receptor complex occurs at the recognition site present on the α subunit. However, the β subunit also contributes towards the pharmacological properties of the receptor binding site by modulating the interaction of the ligand with the α subunit at the binding interface that lies between the α (α2, α3, α4, α6) and β (β2 or β4) subunits in heteromeric receptors. The α5 and β3 subunits do not participate in the acetylcholine binding site and are termed accessory subunits. The binding interface in the pentameric complexes composed of only α (α7-α10) nAChR subunits occurs between two α subunits to result in five identical acetylcholine binding sites 9a, 9c.

Extensive studies using a wide variety of approaches now show that multiple receptor subtypes are generated by the association of different α and β subunits, with the primary ones in the CNS being the α4β2* (the asterisk denotes the possible presence of other nAChR subunits in the receptor complex) and α7 subtypes 9c, 10. These nAChRs are widely distributed throughout the brain, and are localized presynaptically and also somato-dendritically on postsynaptic neurons 9. One of their primary functions is to modulate synaptic transmission and plasticity mediated via other neurotransmitter systems with resultant changes in attention, cognition, depression and affect 9, 11.

The α4β2* and α7 nAChRs are also involved in controlling striatal dopamine function 12 with consequent effects on motor and reward-related responses 11, 12b, 13. Receptors containing solely α4β2 subunits are present on both striatal dopaminergic and glutamatergic terminals, with a subpopulation of α4α5β2 nAChRs expressed only on dopaminergic terminals in rats 10a, 10c, 12a. Receptors containing the α4β2 subunits can exist in two different forms, a high sensitivity subtype with a stoichiometry of (α4)2(β2)3 and a low sensitivity receptor with a stoichiometry of (α4)3(β2)2.14 Thus, both varying α4β2* subunit composition and stoichiometry can generate subtypes with a distinct pharmacological and functional characteristics under different treatment conditions 15. Although the role of striatal postsynaptic α7 nAChRs is still unclear, those localized presynaptically on cortical glutamatergic afferents influence striatal dopamine release.

Pentameric complexes of α6β2* nAChR subtypes are uniquely restricted to CNS catecholaminergic neurons including those in the dopaminergic mesolimbic and nigrostriatal pathway. They are present on dopaminergic terminals and thought to be involved in reward, addiction and motor activity 16. The use of various experimental strategies has identified the presence of two major α6β2* nAChRs, the α6β2β3 and α6α4β2β3 subtypes, as well possibly as a small population expressing only α6β2 subunits 9a, 10a, b, 12a, 17.

In summary, the primary nAChR populations in the striatum includ the α4β2, α4α5β2, α6β2β3, α6α4β2β3 and α7 subtypes. These may be expressed pre- and/or post-synaptically on dopaminergic, glutamatergic, GABAergic and/or other neurons, with a primary role of the presynaptic receptors to modulate dopamine release. The observed heterogeneity in subtypes may be important for fine-tuning of striatal dopaminergic function under varying physiological and pathological conditions.

NACHR MODULATION OF NIGROSTRIATAL DOPAMINE FUNCTION

As mentioned earlier, dopamine neurotransmission is regulated by tonically active cholinergic interneurons that act as a pulsed source of acetylcholine to modulate dopamine release via pre- and postsynaptic nAChRs. NAChR activation subsequently changes membrane excitability and initiates a calcium signaling cascade that ultimately results in neurotransmitter release 7a, 12b, 18. This can occur via direct stimulation of nAChRs on striatal dopaminergic terminals or through the stimulation of nAChRs on striatal glutamatergic terminals 10b, 19.

One approach to measure presynaptic nAChR-mediated dopamine release involves the use of striatal synaptosomes and slices preloaded with 3H-dopamine 12a. This technique, coupled with the use of genetically modified mouse models and the α6 selective neurotoxin α-conotoxinMII, has greatly expanded our understanding of the functional role of nAChR subtypes on the dopamine system 20. Several nAChR subtypes modulate striatal dopamine function, including the α4β2, α4α5β2, α4α6β2β3 and α6β2β3 subtypes, with the α4β2* nAChRs responsible for 50-70% of nAChR-mediated dopamine release and the α6β2* nAChRs the remainder 10c, 12a, 16a.

Another advance that has provided invaluable information about the role of nAChRs in regulating dopamine release is fast scan cyclic voltammetry using striatal slices 12b, 21. This technique allows for the real-time assessment of endogenous dopamine release stimulated electrically at biologically relevant frequencies that mimic typical neuronal firing rates. Experimental studies indicate that nicotine has differential effects on dopamine release depending on the neuronal firing rate with a suppression of release at low stimulation frequencies but not under phasic firing conditions 12b, 21. Studies with slices from knockout mice suggest that β2* nAChR primarily modulate striatal dopamine release 21g. The α6β2* subtype mediates ~80% of nAChR-regulated dopamine release at low firing rates, with the α4β2* receptor population regulating the remainder 21a-c, 22.

In vivo studies have also been done to study the role of nAChRs in the control of striatal dopamine levels. These generally involve microdialysis followed by high pressure liquid chromatography to measure dopamine levels. Acute systemic nicotine or local perfusion of nicotine through the microdialysis probe both increase dopamine levels in striatum 23. Studies with α4, α6 and β2 nAChR knockout mice suggest that the α4β2 nAChR plays a major role in regulating striatal dopamine release in vivo 10b, 24.

These combined results indicate that α6β2* and α4β2* nAChRs play a pivotal role in the control of striatal dopaminergic signaling. This knowledge is fundamental for understanding the nature of the dysfunction in neurological disorders such a Parkinson's disease and developing optimal treatment regimens.

Nigrostriatal damage reduces nAChR-mediated dopamine release

Studies in parkinsonian mice, rats and monkeys show that striatal nAChR subtypes are decreased with striatal dopaminergic denervation 9a, 25. Nigrostriatal degeneration results in differential receptor subtype loss depending on the extent of damage, with a predominant loss of the α6α4β2β3 nAChR with moderate lesioning, a decrease in the α6β2β3 subtype with greater nigrostriatal damage, and a decrease in the α4β2 subtype only with very severe degeneration 17b, 25. Similar results were obtained in brains from Parkinson's disease cases, with 30 to 75% declines in nAChRs in the caudate, putamen and substantia nigra that appear to correlate with the extent of nigrostriatal damage 26. A comparison of changes in α4β2* versus α6β2* nAChRs, showed that greater declines were observed in α6β2* receptors, with the most prominent loss in the α6α4β2β3 as compared to the α6β2β3 nAChR subtype. The α4β2* nAChRs were decreased only with severe degeneration 17b, 17d, 27. Consistent with the animal studies, α7 nAChRs were not changed with nigrostriatal damage. This disparate receptor subtype loss raises the intriguing possibility that the different nAChR subtypes are present on different neuronal populations some of which are more susceptible to neurodegenerative processes than others.

The finding that striatal nAChR subtypes and their function are differentially vulnerable to nigrostriatal damage supports the notion that the different nAChR subtypes are present on different neuronal populations some of which are more susceptible to neurodegenerative processes than others. Such an idea is also supported by the results of cyclic voltammetric studies, which show that there is a different distribution of nAChRs on dopaminergic fibers with various electrophysiological characteristics 21b. For instance, evoked dopamine release from fibers with a low action potential threshold is modulated primarily by α6β2* nAChRs. By contrast, evoked released is influenced predominantly by α4β2* nAChRs for fibers exhibiting higher action potential thresholds. The presence of varying nAChR subtypes on these different classes of striatal fibers may make them differentially vulnerable to neurotoxic agents. Continued studies are in progress to investigate this hypothesis.

The question now arises whether there might be a differential regulation of striatal dopamine release with the preferential loss of one or other nAChR subtype. In rodent striatum, nAChR stimulated dopamine release is decreased with dopaminergic degeneration, as expected. In this species, there was a very close correspondence between the decline in dopamine release and the loss of nAChR receptors 28. Results also suggested that dopamine release mediated by the α4β2* and α6β2* nAChR subpopulations are altered in a corresponding fashion by nigrostriatal damage 28. These data contrast with those obtained in a similar series of experiments in nonhuman primates. Although MPTP-treatment induces similar decreases in nAChRs in monkey and rodent striatum, this decline in receptor density was not associated with a concomitant decrease in either α4β2* or α6β2* mediated dopamine release with moderate nigrostriatal damage 29. These data suggest that there is presynaptic compensation in the release process of monkeys with nigrostriatal damage that involves both the α4β2* and α6β2* nAChR population. These findings are also supported by voltammetry studies that show increased dopamine release in primates after moderate nigrostriatal damage 21d.

Overall, these data show that nAChRs influence dopamine release under control conditions and in the presence of nigrostriatal damage. The primary subtypes involved are the α4β2* and α6β2* nAChR which may be present on different populations of dopaminergic neurons. Their presence on select neuronal populations may play a part in the differential sensitivity of various dopaminergic neurons to nigrostriatal damage. Notably, there appear to be differences in the nAChR mediated regulation of dopamine release in rodents and primates with compensation after moderate damage in nonhuman primates but not rodents. Continued studies in different parkinsonian animal models are essential for a clear understanding of the role of nAChRs in controlling dopamine function in Parkinson's disease.

Chronic nicotine treatment alters nAChR-mediated release

As mentioned, accumulating work suggests that nicotine treatment may be of benefit in Parkinson's disease since it reduces levodopa-induced dyskinesias in parkinsonian animal models and may also play a neuroprotective role against nigrostriatal damage. An important question is thus how chronic nicotine treatment may affect striatal dopamine function. Numerous studies have shown that long term nicotine administration upregulates α4β2* nAChR expression in rodents, non-human primates and human brain 29a, 30. This receptor increase is linked to a concomitant increase in nAChR-mediated dopamine release as measured in brain slices or by microdialysis 31, although a decrease or no change was observed when measured in striatal synaptosomes 30a, 32. This apparent discrepancy now appears to be due to the fact that the enhanced α4β2* receptor expression is accompanied by a downregulation of α6β2* nAChRs such that there is no net change in nAChR mediated dopamine release 30a, 32a, 32d, e.

Some of these results in striatal synaptosomes are supported by our recent cyclic voltammetry studies in striatal slices from rats and primates. In rats, chronic nicotine treatment enhanced nonburst and burst stimulated endogenous dopamine release similar to the overall increase in nAChR-mediated dopamine release in synaptosomes 22. Interestingly, long term nicotine treatment also attenuated the α6β2* nAChR-mediated regulation of striatal dopaminergic function with burst firing, an observation consistent with the nicotine-induced decline in α6β2* nAChR expression and function 22. In contrast, in monkeys, chronic nicotine administration did not enhance either nonburst or burst release and attenuated both α4β2* and α6β2* nAChR-mediated responsiveness in some although not all striatal regions 21c.

These voltammetric data demonstrate that chronic nicotine exposure modulates nAChR-mediated dopamine release in striatum, although the precise functional implications of these cellular changes remain to be elucidated. Greater knowledge of the alterations in nAChR expression and function is important for a clear understanding of the effect of drug treatment under varying pathological conditions such as Parkinson's disease 25.

POTENTIAL ROLE OF NICOTINIC RECEPTOR ACTIVATION IN NIGROSTRIATAL FUNCTION

The morphological overlap coupled with the functional interaction between the dopaminergic and nicotinic cholinergic system provides the biological basis for nicotine's potential to influence behaviors linked to Parkinson's disease. These include the ability of nicotine (1) to reduce levodopa-induced dyskinesias, a debilitating side effect of levodopa therapy, (2) to improve motor symptoms in Parkinson's disease and (3) to protect against nigrostriatal damage. Evidence for such roles for nicotine and the potential importance to Parkinson's disease therapeutics is discussed below.

Nicotine treatment reduces levodopa-induced dyskinesias

Experimental evidence from several animal models now suggests that nicotine may be useful for improving levodopa-induced dyskinesias in Parkinson's disease. These are abnormal involuntary movements that arise with levodopa treatment, the gold-standard for Parkinson's disease therapy. They may be quite mild or so problematic that they compromise the antiparkinsonian effectiveness of levodopa 1, 33. They may develop very rapidly (months) and or more slowly such that the majority of patients develop dyskinesias within 5-10 years 34. Dyskinesias are extremely difficult to prevent. One strategy to postpone their onset involves the initial use of low dose levodopa, but since Parkinson's disease generally progresses with time, increasing doses of levodopa are inevitably required to manage symptoms 1, 33. Dopamine agonists are also less prone to inducing dyskinesias; however they are also less effective at controlling motor symptoms, less well-tolerated overall, and associated with psychiatric and other adverse effects. Drug treatments for reducing levodopa-induced dyskinesias are very limited, with amantadine currently the only accepted pharmacologic approach for reducing their occurrence, although its effects are modest 35. A host of drugs influencing the dopaminergic, serotonergic, glutamatergic, adrenergic, cholinergic, opioid, adenosine and various peptidergic systems are being/have also been tested but in general the drugs tested appear to have limited efficacy 2b, 4, 35b, 36. Additional therapies to reduce levodopa-induced dyskinesias are therefore critical.

Our recent studies show that drugs interacting with the nicotinic cholinergic system are effective in attenuating levodopa-induced dyskinesias. Nicotine, a drug that interacts with multiple nAChR subtypes, reduces levodopa-induced abnormal involuntary movements (AIMs) in three different parkinsonian animal models. Nicotine significantly attenuated dyskinesias in levodopa-treated MPTP-lesioned monkeys, when given either before the onset of dyskinesias or when they were established 3a. We obtained similar results in parkinsonian rodent models of levodopa-induced dyskinesias, attesting to the robustness of the effect of nicotine across species. Nicotine given via several modes of administration (drinking water, minipump or injection) significantly improved levodopa-induced AIMs in rat and mouse parkinsonian models (Bordia et al., 2008). These modes of administration readily lend themselves to use in Parkinson's disease patients, for instance, as an oral formulation or a slow release mode (nicotine patch). Notably, nicotine did not modify the anti-parkinsonian effect of levodopa in any species. This basic work has led to the initiation of a clinical trial to test nicotine against levodopa-induced dyskinesias in Parkinson's disease patients.

The mechanisms whereby nicotine reduces levodopa-induced dyskinetic-like movements are not yet know. However, studies with the nAChR blocker mecamylamine show it involves an interaction at nAChRs 37. The select nAChR populations remain to be elucidated, although the α4β2* and α6β2* subtypes are probably important since these are the primary ones involved in striatal function 9c, 12, 38. Studies with agonists that interact with both α4β2* and α6β2* nAChRs show that such drugs reduce levodopa-induced AIMs almost as effectively as the nonselective nAChR agonist nicotine 39. Studies are currently in progress to identify the role of the α4β2* versus α6β2* nAChR subtypes and the mechanisms whereby an interaction at nAChRs reduces levodopa-induced dyskinesias.

Nicotine and Parkinson's disease motor symptoms

Another question that arises is whether nicotine may improve motor symptoms in Parkinson's disease patients since it is well known to stimulate striatal dopamine release 7a, 12b, 18. Work to address this issue shows that nicotine improves parkinsonian symptoms in ~50% of reports/trials, with improvement in five, no effect in four and a worsening in one 4. The reason for these differences among studies may relate to variations in the mode of administration of nicotine (patch, gum, intravenous), inadequate dosing, timing or duration (days to weeks) of treatment, differences in the degree of parkinsonism and type of trial (open-label versus double-blinded) 40. Work in parkinsonian rats and monkeys show that chronic nicotine treatment in the drinking water or via minipump did not lead to a reduction in parkinsonism either on or off Levodopa 3a, b. These findings in animal models support the results of the trials demonstrating no beneficial effect of nicotine on Parkinson's disease symptoms.

Continued work is necessary to ascertain whether or not nicotine improves Parkinson's disease motor symptoms. Overall, however, these data indicate that nicotine and nicotinic agonists do not worsen, and possibly improve, motor symptoms if such drugs were used for the treatment of levodopa-induced dyskinesias in Parkinson's disease.

Nicotine and neuroprotection

In addition to its effects on Parkinson's disease motor symptoms and dyskinesias, nicotine may also have a long term beneficial action by protecting against nigrostriatal damage. Evidence for such a possibility initially stemmed from the results of epidemiological studies. These showed that a reduced incidence of Parkinson's disease is associated with smoking, in contrast to the enhanced risk associated with other factors such as age, gender, and pesticide exposure 4, 6, 41. An average 50% lower incidence of Parkinson's disease was observed for those who smoked ~20 years prior to disease diagnosis. Moreover, there appeared to be a dose-response relationship, with decreasing Parkinson's disease risk correlated with increasing pack-years smoked.

The putative neuroprotective effect of smoking has been attributed to the ability of nicotine in tobacco to attenuate nigrostriatal data. This possibility is based on results of both in vitro and in vivo experimental studies 5b, 31b, 42. For instance, nicotine protects primary cultured neurons against MPTP or LPS-induced toxicity 42c, d. Moreover, chronic nicotine pretreatment improves neurotoxin-induced nigrostriatal damage in rodents and nonhuman primates, with protection dependent on the extent of neuronal damage and nicotine dosing regimen 31b, 42a, b, 42e, f. As mentioned earlier, the etiology of Parkinson's disease is currently uncertain, with a potential role for both genetic and environmental factors. Experiments done to date investigating nicotine-mediated protection have all involved neurotoxic-induced animal models of nigrostriatal damage that may represent an environmental model of Parkinson's disease. A question that arises is whether nicotine also protects against slowly progressive genetic animal models of Parkinson's disease, such as those involving α-synuclein or other mutations 43. This is an important issue that awaits further study.

In the above in vivo studies nicotine was given prior to and during the development of nigrostriatal damage, a protocol which provides for an assessment of the neuroprotective potential of nicotine. However, epidemiological studies show that once the disease is diagnosed, smoking does not appear to improve Parkinson's disease 44. These latter findings raise the question whether nicotine is only neuroprotective or whether it can also restore function of damaged neurons. Our recent experimental studies in parkinsonian rats and monkeys show that nicotine had no beneficial effect when administrated after nigrostriatal damage is complete. Thus, nicotine's primary role appears to be neuroprotective and not neurorestorative 42b.

The mechanisms underlying nAChR-mediated neuroprotection are currently being investigated. Accumulating evidence suggests that nicotine acts via β2* and α7 nAChRs. The β2* nAChRs, which include the α4β2* and α6β2* subtypes, are expressed on striatal dopamine terminals and appear to be the primary populations involved in protection 42a, b, 45. Further work has identified two α6β2* receptor subpopulations, the α6β2β3 and α6α4β2β3 subtypes, with the latter the most susceptible to neurodegeneration 17b, 42b. Interestingly, the α6α4β2β3 subtype is not decreased in parkinsonian compared to sham-lesioned rats with nicotine treatment. This finding suggests that the presence of the α6α4β2β3 subtype may confer a protective action against nigrostriatal damage 42b. Studies using cultured nigral dopaminergic neurons also suggest that α7 nAChRs present may be important in neuroprotection by modifying immune responsiveness 42d, 46. Taking together, the heteromeric α4β2* nAChR and/or homomeric α7 nAChR appear to contribute to nAChR-mediated neuroprotective effects against nigrostriatal damage.

NAChR activation may mediate neuroprotection by triggering diverse signaling pathways, with the first step most likely involving alterations in cytoplasmic calcium dynamics 47. Elevated intracellular calcium may subsequently activate kinases involved in modulating apoptosis, immune modulator and neurotrophic factor expression 42g, 48. Nicotine decreases the level or activity of pro-apoptotic factors such as caspases and JNK kinases and increase the activity of anti-apoptotic factors 46a, 49. In addition, nicotine treatment enhances FGF-2 and NGF in cell culture, as well as in various brain regions in rats. Altered apoptotic mechanisms coupled with enhanced neurotrophic factor expression may promote survival and protect damaged dopaminergic neurons 21g, 50. Accumulating evidence suggests that inflammatory processes participate in the cascades leading to neuronal degeneration in Parkinson's disease 51. Nicotine has been shown to attenuate immune responses within various brain regions via receptors expressed on microglia and astrocytes 52. Lastly, dopamine released in response to nAChR activation may compete with endogenous or exogenous toxic agents for entry into the dopamine terminal, and thus attenuate neurodegenerative effects.

A point of note is that this reduced disease incidence with smoking appears specific for Parkinson's disease. Similar epidemiological approaches find inconsistent declines in Alzheimer's disease with smoking 53. These discrepancies may be related to the minimum age at study entry with a relative rate for smokers versus nonsmokers ranged from 0.27 to 2.72 for Alzheimer's disease with age (55 to 75 years) 53a. The cellular/molecular basis for protection against nigrostriatal damage as occurs in Parkinson's disease is not known; however, the selective protection against Parkinson's disease may suggest that it involves an interaction between the nicotinic and dopaminergic systems as discussed above.

To conclude, nicotine is neuroprotective although not neurorestorative against nigrostriatal damage, most likely via an interaction at nAChRs. The receptors implicated in this neuroprotection appear to involve the α6β2* and α4β2* subtypes, as well as the α7 receptors. Stimulation of these different subtypes by nicotine may then result in neuroprotection by modulating a host of downstream signaling pathways.

CONCLUSION

Converging evidence suggests that nicotine and/or nicotinic agonists may prove beneficial in the management of Parkinson's disease. This includes their use in the treatment of levodopa-induced dyskinesias, a debilitating side effect that arises in the majority of treated patients. Nicotine may also reduce Parkinson's disease symptoms, although definitive proof awaits further study. Lastly, nicotine treatment may slow down Parkinson's disease progression. This neuroprotection is of particular importance as this implies that nicotine administration may reduce the need for symptomatic treatments.

ACKNOWLEDGEMENTS

This work was supported by NIH grants NS42091, NS47162, NS59910 and the California TRDRP grants 17RT-0119 and 18FT-0058A.

ABBREVIATIONS

AIMs

abnormal involuntary movements

nAChR

nicotinic acetylcholine receptor

*

indicates the possible presence of other subunits in the receptor complex

REFERENCES

  • 1.a Schapira AH, Emre M, Jenner P, Poewe W. Levodopa in the treatment of Parkinson's disease. Eur J Neurol. 2009;16:982–9. doi: 10.1111/j.1468-1331.2009.02697.x. [DOI] [PubMed] [Google Scholar]; b Fahn S. How do you treat motor complications in Parkinson's disease: Medicine, surgery, or both? Ann Neurol. 2009;64(S2):S56–S64. doi: 10.1002/ana.21453. [DOI] [PubMed] [Google Scholar]; c Stocchi F, Tagliati M, Olanow CW. Treatment of levodopa-induced motor complications. Mov Disord. 2008;23(Suppl 3):S599–612. doi: 10.1002/mds.22052. [DOI] [PubMed] [Google Scholar]; d Kieburtz K. Therapeutic strategies to prevent motor complications in Parkinson's disease. J Neurol. 2008;255(Suppl 4):42–5. doi: 10.1007/s00415-008-4007-4. [DOI] [PubMed] [Google Scholar]
  • 2.a Fox SH, Lang AE. Levodopa-related motor complications--phenomenology. Mov Disord. 2008;23(Suppl 3):S509–14. doi: 10.1002/mds.22021. [DOI] [PubMed] [Google Scholar]; b Fox SH, Chuang R, Brotchie JM. Parkinson's disease--opportunities for novel therapeutics to reduce the problems of levodopa therapy. Prog Brain Res. 2008;172:479–94. doi: 10.1016/S0079-6123(08)00923-0. [DOI] [PubMed] [Google Scholar]
  • 3.a Quik M, Cox H, Parameswaran N, O'Leary K, Langston JW, Di Monte D. Nicotine reduces levodopa-induced dyskinesias in lesioned monkeys. Annals of Neurology. 2007;62:588–96. doi: 10.1002/ana.21203. [DOI] [PubMed] [Google Scholar]; b Bordia T, Campos C, Huang L, Quik M. Continuous and intermittent nicotine treatment reduces L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesias in a rat model of Parkinson's disease. The Journal of pharmacology and experimental therapeutics. 2008;327(1):239–47. doi: 10.1124/jpet.108.140897. [DOI] [PubMed] [Google Scholar]; c Huang LZ, Bordia T, Quik M. Nicotine treatment reduces L-dopa-induced dyskinetic-like movements in parkinsonian mice. Society for Neuroscience Abstracts. 2009 on line. [Google Scholar]
  • 4.Quik M, O'Leary K, Tanner CM. Nicotine and Parkinson's disease: implications for therapy. Mov Disord. 2008;23(12):1641–52. doi: 10.1002/mds.21900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.a 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. 2008;84:329–42. doi: 10.1016/j.pneurobio.2007.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]; b O'Neill MJ, Murray TK, Lakics V, Visanji NP, Duty S. The role of neuronal nicotinic acetylcholine receptors in acute and chronic neurodegeneration. Curr Drug Targets CNS Neurol Disord. 2002;1(4):399–411. doi: 10.2174/1568007023339166. [DOI] [PubMed] [Google Scholar]
  • 6.a Elbaz A, Moisan F. Update in the epidemiology of Parkinson's disease. Curr Opin Neurol. 2008;21(4):454–60. doi: 10.1097/WCO.0b013e3283050461. [DOI] [PubMed] [Google Scholar]; b Ritz B, Ascherio A, Checkoway H, Marder KS, Nelson LM, Rocca WA, Ross GW, Strickland D, Van Den Eeden SK, Gorell J. Pooled analysis of tobacco use and risk of Parkinson disease. Arch Neurol. 2007;64(7):990–7. doi: 10.1001/archneur.64.7.990. [DOI] [PubMed] [Google Scholar]; c Thacker EL, O'Reilly EJ, Weisskopf MG, Chen H, Schwarzschild MA, McCullough ML, Calle EE, Thun MJ, Ascherio A. Temporal relationship between cigarette smoking and risk of Parkinson disease. Neurology. 2007;68(10):764–8. doi: 10.1212/01.wnl.0000256374.50227.4b. [DOI] [PMC free article] [PubMed] [Google Scholar]; d Allam MF, Campbell MJ, Hofman A, Del Castillo AS, Fernandez-Crehuet Navajas R. Smoking and Parkinson's disease: systematic review of prospective studies. Mov Disord. 2004;19(6):614–21. doi: 10.1002/mds.20029. [DOI] [PubMed] [Google Scholar]
  • 7.a Zhou FM, Liang Y, Dani JA. Endogenous nicotinic cholinergic activity regulates dopamine release in the striatum. Nat Neurosci. 2001;4(12):1224–9. doi: 10.1038/nn769. [DOI] [PubMed] [Google Scholar]; b Zhou FM, Wilson C, Dani JA. Muscarinic and nicotinic cholinergic mechanisms in the mesostriatal dopamine systems. Neuroscientist. 2003;9(1):23–36. doi: 10.1177/1073858402239588. [DOI] [PubMed] [Google Scholar]
  • 8.a Fuxe K, Hoekfelt T, Nilsson O. Observations on the Cellular Localization of Dopamine in the Caudate Nucleus of the Rat. Z Zellforsch Mikrosk Anat. 1964;63:701–6. doi: 10.1007/BF00339917. [DOI] [PubMed] [Google Scholar]; b Fonnum F. Recent developments in biochemical investigations of cholinergic transmission. Brain Res. 1973;62(2):497–507. doi: 10.1016/0006-8993(73)90714-2. [DOI] [PubMed] [Google Scholar]
  • 9.a Gotti C, Clementi F, Fornari A, Gaimarri A, Guiducci S, Manfredi I, Moretti M, Pedrazzi P, Pucci L, Zoli M. Structural and functional diversity of native brain neuronal nicotinic receptors. Biochem Pharmacol. 2009;78(7):703–11. doi: 10.1016/j.bcp.2009.05.024. [DOI] [PubMed] [Google Scholar]; b Albuquerque EX, Pereira EF, Alkondon M, Rogers SW. Mammalian nicotinic acetylcholine receptors: from structure to function. Physiol Rev. 2009;89(1):73–120. doi: 10.1152/physrev.00015.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Millar NS, Gotti C. Diversity of vertebrate nicotinic acetylcholine receptors. Neuropharmacology. 2009;56(1):237–46. doi: 10.1016/j.neuropharm.2008.07.041. [DOI] [PubMed] [Google Scholar]
  • 10.a Zoli M, Moretti M, Zanardi A, McIntosh JM, Clementi F, Gotti C. Identification of the nicotinic receptor subtypes expressed on dopaminergic terminals in the rat striatum. J Neurosci. 2002;22(20):8785–9. doi: 10.1523/JNEUROSCI.22-20-08785.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Champtiaux N, Gotti C, Cordero-Erausquin M, David DJ, Przybylski C, Lena C, Clementi F, Moretti M, Rossi FM, Le Novere N, McIntosh JM, Gardier AM, Changeux JP. Subunit composition of functional nicotinic receptors in dopaminergic neurons investigated with knockout mice. J Neurosci. 2003;23(21):7820–9. doi: 10.1523/JNEUROSCI.23-21-07820.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Salminen O, Murphy KL, McIntosh JM, Drago J, Marks MJ, Collins AC, Grady SR. Subunit composition and pharmacology of two classes of striatal presynaptic nicotinic acetylcholine receptors mediating dopamine release in mice. Mol Pharmacol. 2004;65(6):1526–35. doi: 10.1124/mol.65.6.1526. [DOI] [PubMed] [Google Scholar]; d Quik M, Vailati S, Bordia T, Kulak JM, Fan H, McIntosh JM, Clementi F, Gotti C. Subunit composition of nicotinic receptors in monkey striatum: effect of treatments with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine or L-DOPA. Mol Pharmacol. 2005;67(1):32–41. doi: 10.1124/mol.104.006015. [DOI] [PubMed] [Google Scholar]
  • 11.Benowitz NL. Pharmacology of nicotine: addiction, smoking-induced disease, and therapeutics. Annual review of pharmacology and toxicology. 2009;49:57–71. doi: 10.1146/annurev.pharmtox.48.113006.094742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.a Grady SR, Salminen O, Laverty DC, Whiteaker P, McIntosh JM, Collins AC, Marks MJ. The subtypes of nicotinic acetylcholine receptors on dopaminergic terminals of mouse striatum. Biochem Pharmacol. 2007;74(8):1235–46. doi: 10.1016/j.bcp.2007.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Exley R, Cragg SJ. Presynaptic nicotinic receptors: a dynamic and diverse cholinergic filter of striatal dopamine neurotransmission. Br J Pharmacol. 2008;153(Suppl 1):S283–97. doi: 10.1038/sj.bjp.0707510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Calabresi P, Di Filippo M. ACh/dopamine crosstalk in motor control and reward: a crucial role for alpha 6-containing nicotinic receptors? Neuron. 2008;60(1):4–7. doi: 10.1016/j.neuron.2008.09.031. [DOI] [PubMed] [Google Scholar]
  • 14.a Nelson ME, Kuryatov A, Choi CH, Zhou Y, Lindstrom J. Alternate stoichiometries of alpha4beta2 nicotinic acetylcholine receptors. Mol Pharmacol. 2003;63(2):332–41. doi: 10.1124/mol.63.2.332. [DOI] [PubMed] [Google Scholar]; b Grady SR, Salminen O, McIntosh JM, Marks MJ, Collins AC. Mouse Striatal Dopamine Nerve Terminals Express alpha4alpha5beta2 and Two Stoichiometric Forms of alpha4beta2*-Nicotinic Acetylcholine Receptors. J Mol Neurosci. 2009;40:91–5. doi: 10.1007/s12031-009-9263-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.a Moroni M, Zwart R, Sher E, Cassels BK, Bermudez I. alpha4beta2 nicotinic receptors with high and low acetylcholine sensitivity: pharmacology, stoichiometry, and sensitivity to long-term exposure to nicotine. Mol Pharmacol. 2006;70(2):755–68. doi: 10.1124/mol.106.023044. [DOI] [PubMed] [Google Scholar]; b Lester HA, Xiao C, Srinivasan R, Son CD, Miwa J, Pantoja R, Banghart MR, Dougherty DA, Goate AM, Wang JC. Nicotine is a selective pharmacological chaperone of acetylcholine receptor number and stoichiometry. Implications for drug discovery. Aaps J. 2009;11(1):167–77. doi: 10.1208/s12248-009-9090-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.a Drenan RM, Grady SR, Whiteaker P, McClure-Begley T, McKinney S, Miwa JM, Bupp S, Heintz N, McIntosh JM, Bencherif M, Marks MJ, Lester HA. In vivo activation of midbrain dopamine neurons via sensitized, high-affinity alpha 6 nicotinic acetylcholine receptors. Neuron. 2008;60(1):123–36. doi: 10.1016/j.neuron.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Pons S, Fattore L, Cossu G, Tolu S, Porcu E, McIntosh JM, Changeux JP, Maskos U, Fratta W. Crucial role of alpha4 and alpha6 nicotinic acetylcholine receptor subunits from ventral tegmental area in systemic nicotine self-administration. J Neurosci. 2008;28(47):12318–27. doi: 10.1523/JNEUROSCI.3918-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.a Jones IW, Bolam JP, Wonnacott S. Presynaptic localisation of the nicotinic acetylcholine receptor beta2 subunit immunoreactivity in rat nigrostriatal dopaminergic neurones. J Comp Neurol. 2001;439(2):235–47. doi: 10.1002/cne.1345. [DOI] [PubMed] [Google Scholar]; b Bordia T, Grady SR, McIntosh JM, Quik M. Nigrostriatal damage preferentially decreases a subpopulation of alpha6beta2* nAChRs in mouse, monkey, and Parkinson's disease striatum. Mol Pharmacol. 2007;72(1):52–61. doi: 10.1124/mol.107.035998. [DOI] [PubMed] [Google Scholar]; c Champtiaux N, Han ZY, Bessis A, Rossi FM, Zoli M, Marubio L, McIntosh JM, Changeux JP. Distribution and pharmacology of alpha 6-containing nicotinic acetylcholine receptors analyzed with mutant mice. J Neurosci. 2002;22(4):1208–17. doi: 10.1523/JNEUROSCI.22-04-01208.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]; d Quik M, Bordia T, Forno L, McIntosh JM. Loss of alpha-conotoxinMII- and A85380-sensitive nicotinic receptors in Parkinson's disease striatum. J Neurochem. 2004;88(3):668–79. doi: 10.1111/j.1471-4159.2004.02177.x. [DOI] [PubMed] [Google Scholar]
  • 18.a Dajas-Bailador F, Wonnacott S. Nicotinic acetylcholine receptors and the regulation of neuronal signalling. Trends Pharmacol Sci. 2004;25(6):317–24. doi: 10.1016/j.tips.2004.04.006. [DOI] [PubMed] [Google Scholar]; b Livingstone PD, Wonnacott S. Nicotinic acetylcholine receptors and the ascending dopamine pathways. Biochem Pharmacol. 2009;78(7):744–55. doi: 10.1016/j.bcp.2009.06.004. [DOI] [PubMed] [Google Scholar]
  • 19.a Marchi M, Risso F, Viola C, Cavazzani P, Raiteri M. Direct evidence that release-stimulating alpha7* nicotinic cholinergic receptors are localized on human and rat brain glutamatergic axon terminals. J Neurochem. 2002;80(6):1071–8. doi: 10.1046/j.0022-3042.2002.00805.x. [DOI] [PubMed] [Google Scholar]; b Wooltorton JR, Pidoplichko VI, Broide RS, Dani JA. Differential desensitization and distribution of nicotinic acetylcholine receptor subtypes in midbrain dopamine areas. J Neurosci. 2003;23(8):3176–85. doi: 10.1523/JNEUROSCI.23-08-03176.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.a Azam L, McIntosh JM. Alpha-conotoxins as pharmacological probes of nicotinic acetylcholine receptors. Acta Pharmacol Sin. 2009;30(6):771–83. doi: 10.1038/aps.2009.47. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Kulak JM, Nguyen TA, Olivera BM, McIntosh JM. Alpha-conotoxin MII blocks nicotine-stimulated dopamine release in rat striatal synaptosomes. J Neurosci. 1997;17(14):5263–70. doi: 10.1523/JNEUROSCI.17-14-05263.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.a Exley R, Clements MA, Hartung H, McIntosh JM, Cragg SJ. Alpha6-containing nicotinic acetylcholine receptors dominate the nicotine control of dopamine neurotransmission in nucleus accumbens. Neuropsychopharmacology. 2008;33(9):2158–66. doi: 10.1038/sj.npp.1301617. [DOI] [PubMed] [Google Scholar]; b Meyer EL, Yoshikami D, McIntosh JM. The neuronal nicotinic acetylcholine receptors alpha 4* and alpha 6* differentially modulate dopamine release in mouse striatal slices. J Neurochem. 2008;105(5):1761–9. doi: 10.1111/j.1471-4159.2008.05266.x. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Perez XA, O'Leary KT, Parameswaran N, McIntosh JM, Quik M. Prominent role of alpha3/alpha6beta2* nAChRs in regulating evoked dopamine release in primate putamen: effect of long-term nicotine treatment. Mol Pharmacol. 2009;75(4):938–46. doi: 10.1124/mol.108.053801. [DOI] [PMC free article] [PubMed] [Google Scholar]; d Perez XA, Parameswaran N, Huang LZ, O'Leary KT, Quik M. Pre-synaptic dopaminergic compensation after moderate nigrostriatal damage in non-human primates. J Neurochem. 2008;105(5):1861–72. doi: 10.1111/j.1471-4159.2008.05268.x. [DOI] [PMC free article] [PubMed] [Google Scholar]; e Rice ME, Cragg SJ. Nicotine amplifies reward-related dopamine signals in striatum. Nat Neurosci. 2004;7(6):583–4. doi: 10.1038/nn1244. [DOI] [PubMed] [Google Scholar]; f Zhang L, Zhou FM, Dani JA. Cholinergic drugs for Alzheimer's disease enhance in vitro dopamine release. Mol Pharmacol. 2004;66(3):538–44. doi: 10.1124/mol.104.000299. [DOI] [PubMed] [Google Scholar]; g Zhang T, Zhang L, Liang Y, Siapas AG, Zhou FM, Dani JA. Dopamine signaling differences in the nucleus accumbens and dorsal striatum exploited by nicotine. J Neurosci. 2009;29(13):4035–43. doi: 10.1523/JNEUROSCI.0261-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]; h Zhang X, Zhou Z, Wang D, Li A, Yin Y, Gu X, Ding F, Zhen X, Zhou J. Activation of phosphatidylinositol-linked D1-like receptor modulates FGF-2 expression in astrocytes via IP3-dependent Ca2+ signaling. J Neurosci. 2009;29(24):7766–75. doi: 10.1523/JNEUROSCI.0389-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.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(3):844–53. doi: 10.1124/mol.108.048843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.a Brazell MP, Mitchell SN, Joseph MH, Gray JA. Acute administration of nicotine increases the in vivo extracellular levels of dopamine, 3,4-dihydroxyphenylacetic acid and ascorbic acid preferentially in the nucleus accumbens of the rat: comparison with caudate-putamen. Neuropharmacology. 1990;29(12):1177–85. doi: 10.1016/0028-3908(90)90042-p. [DOI] [PubMed] [Google Scholar]; b Domino EF, Tsukada H. Nicotine sensitization of monkey striatal dopamine release. Eur J Pharmacol. 2009;607(1-3):91–5. doi: 10.1016/j.ejphar.2009.02.011. [DOI] [PubMed] [Google Scholar]; c Imperato A, Mulas A, Di Chiara G. Nicotine preferentially stimulates dopamine release in the limbic system of freely moving rats. Eur J Pharmacol. 1986;132(2-3):337–8. doi: 10.1016/0014-2999(86)90629-1. [DOI] [PubMed] [Google Scholar]; d Lecca D, Shim I, Costa E, Javaid JI. Striatal application of nicotine, but not of lobeline, attenuates dopamine release in freely moving rats. Neuropharmacology. 2000;39(1):88–98. doi: 10.1016/s0028-3908(99)00085-4. [DOI] [PubMed] [Google Scholar]; e Marshall DL, Redfern PH, Wonnacott S. Presynaptic nicotinic modulation of dopamine release in the three ascending pathways studied by in vivo microdialysis: comparison of naive and chronic nicotine-treated rats. J Neurochem. 1997;68(4):1511–9. doi: 10.1046/j.1471-4159.1997.68041511.x. [DOI] [PubMed] [Google Scholar]; f Toth E, Sershen H, Hashim A, Vizi ES, Lajtha A. Effect of nicotine on extracellular levels of neurotransmitters assessed by microdialysis in various brain regions: role of glutamic acid. Neurochem Res. 1992;17(3):265–71. doi: 10.1007/BF00966669. [DOI] [PubMed] [Google Scholar]
  • 24.a Marubio LM, Gardier AM, Durier S, David D, Klink R, Arroyo-Jimenez MM, McIntosh JM, Rossi F, Champtiaux N, Zoli M, Changeux JP. Effects of nicotine in the dopaminergic system of mice lacking the alpha4 subunit of neuronal nicotinic acetylcholine receptors. Eur J Neurosci. 2003;17(7):1329–37. doi: 10.1046/j.1460-9568.2003.02564.x. [DOI] [PubMed] [Google Scholar]; b Picciotto MR, Zoli M, Rimondini R, Lena C, Marubio LM, Pich EM, Fuxe K, Changeux JP. Acetylcholine receptors containing the beta2 subunit are involved in the reinforcing properties of nicotine. Nature. 1998;391(6663):173–7. doi: 10.1038/34413. [DOI] [PubMed] [Google Scholar]
  • 25.Quik M, Huang LZ, Parameswaran N, Bordia T, Campos C, Perez XA. Multiple roles for nicotine in Parkinson's disease. Biochem Pharmacol. 2009;78(7):677–85. doi: 10.1016/j.bcp.2009.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.a Aubert I, Araujo DM, Cecyre D, Robitaille Y, Gauthier S, Quirion R. Comparative alterations of nicotinic and muscarinic binding sites in Alzheimer's and Parkinson's diseases. J Neurochem. 1992;58(2):529–41. doi: 10.1111/j.1471-4159.1992.tb09752.x. [DOI] [PubMed] [Google Scholar]; b Court JA, Piggott MA, Lloyd S, Cookson N, Ballard CG, McKeith IG, Perry RH, Perry EK. Nicotine binding in human striatum: elevation in schizophrenia and reductions in dementia with Lewy bodies, Parkinson's disease and Alzheimer's disease and in relation to neuroleptic medication. Neuroscience. 2000;98(1):79–87. doi: 10.1016/s0306-4522(00)00071-3. [DOI] [PubMed] [Google Scholar]; c Perry EK, Morris CM, Court JA, Cheng A, Fairbairn AF, McKeith IG, Irving D, Brown A, Perry RH. Alteration in nicotine binding sites in Parkinson's disease, Lewy body dementia and Alzheimer's disease: possible index of early neuropathology. Neuroscience. 1995;64(2):385–95. doi: 10.1016/0306-4522(94)00410-7. [DOI] [PubMed] [Google Scholar]; d Rinne JO, Myllykyla T, Lonnberg P, Marjamaki P. A postmortem study of brain nicotinic receptors in Parkinson's and Alzheimer's disease. Brain Res. 1991;547(1):167–70. doi: 10.1016/0006-8993(91)90588-m. [DOI] [PubMed] [Google Scholar]
  • 27.a Bohr IJ, Ray MA, McIntosh JM, Chalon S, Guilloteau D, McKeith IG, Perry RH, Clementi F, Perry EK, Court JA, Piggott MA. Cholinergic nicotinic receptor involvement in movement disorders associated with Lewy body diseases. An autoradiography study using [(125)I]alpha-conotoxinMII in the striatum and thalamus. Exp Neurol. 2005;191(2):292–300. doi: 10.1016/j.expneurol.2004.10.004. [DOI] [PubMed] [Google Scholar]; b Gotti C, Moretti M, Bohr I, Ziabreva I, Vailati S, Longhi R, Riganti L, Gaimarri A, McKeith IG, Perry RH, Aarsland D, Larsen JP, Sher E, Beattie R, Clementi F, Court JA. Selective nicotinic acetylcholine receptor subunit deficits identified in Alzheimer's disease, Parkinson's disease and dementia with Lewy bodies by immunoprecipitation. Neurobiol Dis. 2006;23(2):481–9. doi: 10.1016/j.nbd.2006.04.005. [DOI] [PubMed] [Google Scholar]
  • 28.Quik M, Sum JD, Whiteaker P, McCallum SE, Marks MJ, Musachio J, McIntosh JM, Collins AC, Grady SR. Differential declines in striatal nicotinic receptor subtype function after nigrostriatal damage in mice. Mol Pharmacol. 2003;63(5):1169–79. doi: 10.1124/mol.63.5.1169. [DOI] [PubMed] [Google Scholar]
  • 29.a 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. The Journal of pharmacology and experimental therapeutics. 2006;318(1):381–8. doi: 10.1124/jpet.106.104414. [DOI] [PubMed] [Google Scholar]; b McCallum SE, Parameswaran N, Bordia T, McIntosh JM, Grady SR, Quik M. Decrease in alpha3*/alpha6* nicotinic receptors but not nicotine-evoked dopamine release in monkey brain after nigrostriatal damage. Mol Pharmacol. 2005;68(3):737–46. doi: 10.1124/mol.105.012773. [DOI] [PubMed] [Google Scholar]
  • 30.a McCallum SE, Parameswaran N, Bordia T, Fan H, Tyndale RF, Langston JW, McIntosh JM, Quik M. 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(4):1028–41. doi: 10.1111/j.1471-4159.2005.03646.x. [DOI] [PubMed] [Google Scholar]; b Marks MJ, Burch JB, Collins AC. Effects of chronic nicotine infusion on tolerance development and nicotinic receptors. J Pharmacol Exp Ther. 1983;226(3):817–25. [PubMed] [Google Scholar]; c Schwartz RD, Kellar KJ. Nicotinic cholinergic receptor binding sites in the brain: regulation in vivo. Science. 1983;220(4593):214–6. doi: 10.1126/science.6828889. [DOI] [PubMed] [Google Scholar]; d Benwell ME, Balfour DJ, Anderson JM. Evidence that tobacco smoking increases the density of (-)-[3H]nicotine binding sites in human brain. J Neurochem. 1988;50(4):1243–7. doi: 10.1111/j.1471-4159.1988.tb10600.x. [DOI] [PubMed] [Google Scholar]; e Perry DC, Davila-Garcia MI, Stockmeier CA, Kellar KJ. Increased nicotinic receptors in brains from smokers: membrane binding and autoradiography studies. J Pharmacol Exp Ther. 1999;289(3):1545–52. [PubMed] [Google Scholar]
  • 31.a Rahman S, Zhang J, Corrigall WA. Effects of acute and chronic nicotine on somatodendritic dopamine release of the rat ventral tegmental area: in vivo microdialysis study. Neurosci Lett. 2003;348(2):61–4. doi: 10.1016/s0304-3940(03)00723-7. [DOI] [PubMed] [Google Scholar]; b Visanji NP, Mitchell SN, O'Neill MJ, Duty S. Chronic pre-treatment with nicotine enhances nicotine-evoked striatal dopamine release and alpha6 and beta3 nicotinic acetylcholine receptor subunit mRNA in the substantia nigra pars compacta of the rat. Neuropharmacology. 2006;50(1):36–46. doi: 10.1016/j.neuropharm.2005.07.013. [DOI] [PubMed] [Google Scholar]; c Yu ZJ, Wecker L. Chronic nicotine administration differentially affects neurotransmitter release from rat striatal slices. J Neurochem. 1994;63(1):186–94. doi: 10.1046/j.1471-4159.1994.63010186.x. [DOI] [PubMed] [Google Scholar]
  • 32.a Grady SR, Grun EU, Marks MJ, Collins AC. Pharmacological comparison of transient and persistent [3H]dopamine release from mouse striatal synaptosomes and response to chronic L-nicotine treatment. The Journal of pharmacology and experimental therapeutics. 1997;282(1):32–43. [PubMed] [Google Scholar]; b Grilli M, Parodi M, Raiteri M, Marchi M. Chronic nicotine differentially affects the function of nicotinic receptor subtypes regulating neurotransmitter release. J Neurochem. 2005;93(5):1353–60. doi: 10.1111/j.1471-4159.2005.03126.x. [DOI] [PubMed] [Google Scholar]; c Jacobs I, Anderson DJ, Surowy CS, Puttfarcken PS. Differential regulation of nicotinic receptor-mediated neurotransmitter release following chronic (-)-nicotine administration. Neuropharmacology. 2002;43(5):847–56. doi: 10.1016/s0028-3908(02)00166-1. [DOI] [PubMed] [Google Scholar]; d Lai A, Parameswaran N, Khwaja M, Whiteaker P, Lindstrom JM, Fan H, McIntosh JM, Grady SR, Quik M. Long-term nicotine treatment decreases striatal alpha 6* nicotinic acetylcholine receptor sites and function in mice. Mol Pharmacol. 2005;67(5):1639–47. doi: 10.1124/mol.104.006429. [DOI] [PubMed] [Google Scholar]; e Marks MJ, Grady SR, Collins AC. Downregulation of nicotinic receptor function after chronic nicotine infusion. The Journal of pharmacology and experimental therapeutics. 1993;266(3):1268–76. [PubMed] [Google Scholar]
  • 33.Lang AE. When and how should treatment be started in Parkinson disease? Neurology. 2009;72(7 Suppl):S39–43. doi: 10.1212/WNL.0b013e318198e177. [DOI] [PubMed] [Google Scholar]
  • 34.Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord. 2001;16(3):448–58. doi: 10.1002/mds.1090. [DOI] [PubMed] [Google Scholar]
  • 35.a Thanvi B, Lo N, Robinson T. Levodopa-induced dyskinesia in Parkinson's disease: clinical features, pathogenesis, prevention and treatment. Postgrad Med J. 2007;83(980):384–8. doi: 10.1136/pgmj.2006.054759. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Fabbrini G, Brotchie JM, Grandas F, Nomoto M, Goetz CG. Levodopa-induced dyskinesias. Mov Disord. 2007 doi: 10.1002/mds.21475. [DOI] [PubMed] [Google Scholar]
  • 36.a Brotchie JM, Lee J, Venderova K. Levodopa-induced dyskinesia in Parkinson's disease. J Neural Transm. 2005;112(3):359–91. doi: 10.1007/s00702-004-0251-7. [DOI] [PubMed] [Google Scholar]; b Cenci MA, Lundblad M. Post- versus presynaptic plasticity in L-DOPA-induced dyskinesia. J Neurochem. 2006;99:381–92. doi: 10.1111/j.1471-4159.2006.04124.x. [DOI] [PubMed] [Google Scholar]; c Cenci MA, Lundblad M. Ratings of L-DOPA-induced dyskinesia in the unilateral 6-OHDA lesion model of Parkinson's disease in rats and mice. Curr Protoc Neurosci. 2007 doi: 10.1002/0471142301.ns0925s41. Chapter 9, Unit 9 25. [DOI] [PubMed] [Google Scholar]; d Guigoni C, Aubert I, Li Q, Gurevich VV, Benovic JL, Ferry S, Mach U, Stark H, Leriche L, Hakansson K, Bioulac BH, Gross CE, Sokoloff P, Fisone G, Gurevich EV, Bloch B, Bezard E. Pathogenesis of levodopa-induced dyskinesia: focus on D1 and D3 dopamine receptors. Parkinsonism Relat Disord. 2005;11(Suppl 1):S25–9. doi: 10.1016/j.parkreldis.2004.11.005. [DOI] [PubMed] [Google Scholar]; e Guigoni C, Li Q, Aubert I, Dovero S, Bioulac BH, Bloch B, Crossman AR, Gross CE, Bezard E. Involvement of sensorimotor, limbic, and associative basal ganglia domains in L-3,4-dihydroxyphenylalanine-induced dyskinesia. J Neurosci. 2005;25(8):2102–7. doi: 10.1523/JNEUROSCI.5059-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]; f Linazasoro G. New ideas on the origin of L-dopa-induced dyskinesias: age, genes and neural plasticity. Trends Pharmacol Sci. 2005;26(8):391–7. doi: 10.1016/j.tips.2005.06.007. [DOI] [PubMed] [Google Scholar]; g Mercuri NB, Bernardi G. The ‘magic’ of L-dopa: why is it the gold standard Parkinson's disease therapy? Trends Pharmacol Sci. 2005;26(7):341–4. doi: 10.1016/j.tips.2005.05.002. [DOI] [PubMed] [Google Scholar]; h Samadi P, Bedard PJ, Rouillard C. Opioids and motor complications in Parkinson's disease. Trends Pharmacol Sci. 2006;27(10):512–7. doi: 10.1016/j.tips.2006.08.002. [DOI] [PubMed] [Google Scholar]; i Linazasoro G, Van Blercom N, Ugedo L, Ruiz Ortega JA. Pharmacological treatment of Parkinson's disease: life beyond dopamine D2/D3 receptors? J Neural Transm. 2008;115(3):431–41. doi: 10.1007/s00702-007-0852-z. [DOI] [PubMed] [Google Scholar]; j Carta M, Carlsson T, Munoz A, Kirik D, Bjorklund A. Serotonin-dopamine interaction in the induction and maintenance of L-DOPA-induced dyskinesias. Prog Brain Res. 2008;172:465–78. doi: 10.1016/S0079-6123(08)00922-9. [DOI] [PubMed] [Google Scholar]
  • 37.Bordia T, Campos C, McIntosh JM, Quik M. Nicotinic receptor-mediated reduction in L-dopa-induced dyskinesias may occur via desensitization. The Journal of pharmacology and experimental therapeutics. 2010;333:929–38. doi: 10.1124/jpet.109.162396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.a Gotti C, Moretti M, Gaimarri A, Zanardi A, Clementi F, Zoli M. Heterogeneity and complexity of native brain nicotinic receptors. Biochem Pharmacol. 2007;74(8):1102–11. doi: 10.1016/j.bcp.2007.05.023. [DOI] [PubMed] [Google Scholar]; b Quik M, Bordia T, O'Leary K. Nicotinic receptors as CNS targets for Parkinson's disease. Biochem Pharmacol. 2007;74:1224–1234. doi: 10.1016/j.bcp.2007.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Huang LZ, Campos C, Ly J, Carroll FI, Quik M. Nicotinic receptor agonists decrease L-dopa-induced dyskinesias most effectively in moderately lesioned parkinsonian rats. Neuropharmacology. 2011;60:861–868. doi: 10.1016/j.neuropharm.2010.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.a Ishikawa A, Miyatake T. Effects of smoking in patients with early-onset Parkinson's disease. J Neurol Sci. 1993;117(1-2):28–32. doi: 10.1016/0022-510x(93)90150-w. [DOI] [PubMed] [Google Scholar]; b Fagerstrom KO, Pomerleau O, Giordani B, Stelson F. Nicotine may relieve symptoms of Parkinson's disease. Psychopharmacology (Berl) 1994;116(1):117–9. doi: 10.1007/BF02244882. [DOI] [PubMed] [Google Scholar]; c Clemens P, Baron JA, Coffey D, Reeves A. The short-term effect of nicotine chewing gum in patients with Parkinson's disease. Psychopharmacology (Berl) 1995;117(2):253–6. doi: 10.1007/BF02245195. [DOI] [PubMed] [Google Scholar]; d Ebersbach G, Stock M, Muller J, Wenning G, Wissel J, Poewe W. Worsening of motor performance in patients with Parkinson's disease following transdermal nicotine administration. Mov Disord. 1999;14(6):1011–3. doi: 10.1002/1531-8257(199911)14:6<1011::aid-mds1016>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]; e Kelton MC, Kahn HJ, Conrath CL, Newhouse PA. The effects of nicotine on Parkinson's disease. Brain Cogn. 2000;43(1-3):274–82. [PubMed] [Google Scholar]; f Vieregge A, Sieberer M, Jacobs H, Hagenah JM, Vieregge P. Transdermal nicotine in PD: a randomized, double-blind, placebo-controlled study. Neurology. 2001;57(6):1032–5. doi: 10.1212/wnl.57.6.1032. [DOI] [PubMed] [Google Scholar]; g Villafane G, Cesaro P, Rialland A, Baloul S, Azimi S, Bourdet C, Le Houezec J, Macquin-Mavier I, Maison P. Chronic high dose transdermal nicotine in Parkinson's disease: an open trial. Eur J Neurol. 2007;14:1313–1316. doi: 10.1111/j.1468-1331.2007.01949.x. [DOI] [PubMed] [Google Scholar]; h Shoulson I. Randomized placebo-controlled study of the nicotinic agonist SIB-1508Y in Parkinson disease. Neurology. 2006;66(3):408–10. doi: 10.1212/01.wnl.0000196466.99381.5c. [DOI] [PubMed] [Google Scholar]; i Lemay S, Chouinard S, Blanchet P, Masson H, Soland V, Beuter A, Bedard MA. Lack of efficacy of a nicotine transdermal treatment on motor and cognitive deficits in Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28(1):31–9. doi: 10.1016/S0278-5846(03)00172-6. [DOI] [PubMed] [Google Scholar]
  • 41.Hancock DB, Martin ER, Mayhew GM, Stajich JM, Jewett R, Stacy MA, Scott BL, Vance JM, Scott WK. Pesticide exposure and risk of Parkinson's disease: a family-based case-control study. BMC Neurol. 2008;8:6. doi: 10.1186/1471-2377-8-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.a Costa G, Abin-Carriquiry JA, Dajas F. Nicotine prevents striatal dopamine loss produced by 6-hydroxydopamine lesion in the substantia nigra. Brain Res. 2001;888(2):336–342. doi: 10.1016/s0006-8993(00)03087-0. [DOI] [PubMed] [Google Scholar]; b Huang LZ, Parameswaran N, Bordia T, Michael McIntosh J, Quik M. Nicotine is neuroprotective when administered before but not after nigrostriatal damage in rats and monkeys. J Neurochem. 2009;109(3):826–37. doi: 10.1111/j.1471-4159.2009.06011.x. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Jeyarasasingam G, Tompkins L, Quik M. Stimulation of non-alpha7 nicotinic receptors partially protects dopaminergic neurons from 1-methyl-4-phenylpyridinium-induced toxicity in culture. Neuroscience. 2002;109(2):275–85. doi: 10.1016/s0306-4522(01)00488-2. [DOI] [PubMed] [Google Scholar]; d Park HJ, Lee PH, Ahn YW, Choi YJ, Lee G, Lee DY, Chung ES, Jin BK. Neuroprotective effect of nicotine on dopaminergic neurons by anti-inflammatory action. Eur J Neurosci. 2007;26(1):79–89. doi: 10.1111/j.1460-9568.2007.05636.x. [DOI] [PubMed] [Google Scholar]; e Quik M, O'Neill M, Perez XA. Nicotine neuroprotection against nigrostriatal damage: importance of the animal model. Trends Pharmacol Sci. 2007;28:229–35. doi: 10.1016/j.tips.2007.03.001. [DOI] [PubMed] [Google Scholar]; f Quik M, Parameswaran N, McCallum SE, Bordia T, Bao S, McCormack A, Kim A, Tyndale RF, Langston JW, Di Monte DA. Chronic oral nicotine treatment protects against striatal degeneration in MPTP-treated primates. J Neurochem. 2006;98(6):1866–75. doi: 10.1111/j.1471-4159.2006.04078.x. [DOI] [PubMed] [Google Scholar]; g Picciotto MR, Zoli M. Neuroprotection via nAChRs: the role of nAChRs in neurodegenerative disorders such as Alzheimer's and Parkinson's disease. Front Biosci. 2008;13:492–504. doi: 10.2741/2695. [DOI] [PubMed] [Google Scholar]
  • 43.a Bazzu G, Calia G, Puggioni G, Spissu Y, Rocchitta G, Debetto P, Grigoletto J, Zusso M, Migheli R, Serra PA, Desole MS, Miele E. alpha-Synuclein- and MPTP-generated rodent models of Parkinson's disease and the study of extracellular striatal dopamine dynamics: a microdialysis approach. CNS Neurol Disord Drug Targets. 2010;9(4):482–90. doi: 10.2174/187152710791556177. [DOI] [PubMed] [Google Scholar]; b Chesselet MF. In vivo alpha-synuclein overexpression in rodents: a useful model of Parkinson's disease? Exp Neurol. 2008;209(1):22–7. doi: 10.1016/j.expneurol.2007.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]; c Ulusoy A, Decressac M, Kirik D, Bjorklund A. Viral vector-mediated overexpression of alpha-synuclein as a progressive model of Parkinson's disease. Prog Brain Res. 2010;184:89–111. doi: 10.1016/S0079-6123(10)84005-1. [DOI] [PubMed] [Google Scholar]; d Mandel SA, Fishman-Jacob T, Youdim MB. Modeling sporadic Parkinson's disease by silencing the ubiquitin E3 ligase component, SKP1A. Parkinsonism Relat Disord. 2009;15(Suppl 3):S148–51. doi: 10.1016/S1353-8020(09)70803-X. [DOI] [PubMed] [Google Scholar]
  • 44.Alves G, Kurz M, Lie SA, Larsen JP. Cigarette smoking in Parkinson's disease: influence on disease progression. Mov Disord. 2004;19(9):1087–1092. doi: 10.1002/mds.20117. [DOI] [PubMed] [Google Scholar]
  • 45.a Khwaja M, McCormack A, McIntosh JM, Di Monte DA, Quik M. Nicotine partially protects against paraquat-induced nigrostriatal damage in mice; link to alpha6beta2* nAChRs. J Neurochem. 2007;100(1):180–90. doi: 10.1111/j.1471-4159.2006.04177.x. [DOI] [PubMed] [Google Scholar]; b Ryan RE, Ross SA, Drago J, Loiacono RE. Dose-related neuroprotective effects of chronic nicotine in 6-hydroxydopamine treated rats, and loss of neuroprotection in alpha4 nicotinic receptor subunit knockout mice. Br J Pharmacol. 2001;132(8):1650–6. doi: 10.1038/sj.bjp.0703989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.a Marrero MB, Bencherif M. Convergence of alpha 7 nicotinic acetylcholine receptor-activated pathways for anti-apoptosis and anti-inflammation: central role for JAK2 activation of STAT3 and NF-kappaB. Brain Res. 2009;1256:1–7. doi: 10.1016/j.brainres.2008.11.053. [DOI] [PubMed] [Google Scholar]; b Takeuchi H, Yanagida T, Inden M, Takata K, Kitamura Y, Yamakawa K, Sawada H, Izumi Y, Yamamoto N, Kihara T, Uemura K, Inoue H, Taniguchi T, Akaike A, Takahashi R, Shimohama S. Nicotinic receptor stimulation protects nigral dopaminergic neurons in rotenone-induced Parkinson's disease models. J Neurosci Res. 2009;87(2):576–85. doi: 10.1002/jnr.21869. [DOI] [PubMed] [Google Scholar]
  • 47.a Ren K, Puig V, Papke RL, Itoh Y, Hughes JA, Meyer EM. Multiple calcium channels and kinases mediate alpha7 nicotinic receptor neuroprotection in PC12 cells. J Neurochem. 2005;94(4):926–33. doi: 10.1111/j.1471-4159.2005.03223.x. [DOI] [PubMed] [Google Scholar]; b Shen JX, Yakel JL. Nicotinic acetylcholine receptor-mediated calcium signaling in the nervous system. Acta Pharmacol Sin. 2009;30(6):673–80. doi: 10.1038/aps.2009.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Quik M. Smoking, nicotine and Parkinson's disease. Trends Neurosci. 2004;27(9):561–8. doi: 10.1016/j.tins.2004.06.008. [DOI] [PubMed] [Google Scholar]
  • 49.a Liu Q, Zhao B. Nicotine attenuates beta-amyloid peptide-induced neurotoxicity, free radical and calcium accumulation in hippocampal neuronal cultures. Br J Pharmacol. 2004;141(4):746–54. doi: 10.1038/sj.bjp.0705653. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Zhao J, Xin M, Wang T, Zhang Y, Deng X. Nicotine Enhances the Antiapoptotic Function of Mcl-1 through Phosphorylation. Mol Cancer Res. 2009 doi: 10.1158/1541-7786.MCR-09-0304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.von Bohlen und Halbach O, Unsicker K. Neurotrophic support of midbrain dopaminergic neurons. Adv Exp Med Biol. 2009;651:73–80. doi: 10.1007/978-1-4419-0322-8_7. [DOI] [PubMed] [Google Scholar]
  • 51.a Luchtman DW, Shao D, Song C. Behavior, neurotransmitters and inflammation in three regimens of the MPTP mouse model of Parkinson's disease. Physiol Behav. 2009;98(1-2):130–8. doi: 10.1016/j.physbeh.2009.04.021. [DOI] [PubMed] [Google Scholar]; b Stolp HB, Dziegielewska KM. Review: Role of developmental inflammation and blood-brain barrier dysfunction in neurodevelopmental and neurodegenerative diseases. Neuropathol Appl Neurobiol. 2009;35(2):132–46. doi: 10.1111/j.1365-2990.2008.01005.x. [DOI] [PubMed] [Google Scholar]
  • 52.Shi FD, Piao WH, Kuo YP, Campagnolo DI, Vollmer TL, Lukas RJ. Nicotinic attenuation of central nervous system inflammation and autoimmunity. J Immunol. 2009;182(3):1730–9. doi: 10.4049/jimmunol.182.3.1730. [DOI] [PubMed] [Google Scholar]
  • 53.a Duron E, Hanon O. Vascular risk factors, cognitive decline, and dementia. Vasc Health Risk Manag. 2008;4(2):363–81. doi: 10.2147/vhrm.s1839. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Sabbagh MN, Lukas RJ, Sparks DL, Reid RT. The nicotinic acetylcholine receptor, smoking, and Alzheimer's disease. J Alzheimers Dis. 2002;4(4):317–25. doi: 10.3233/jad-2002-4407. [DOI] [PubMed] [Google Scholar]

RESOURCES