• Nie Znaleziono Wyników

Nicotine dependence – human and animal studies, current pharmacotherapies and future

N/A
N/A
Protected

Academic year: 2022

Share "Nicotine dependence – human and animal studies, current pharmacotherapies and future"

Copied!
9
0
0

Pełen tekst

(1)

Review

Nicotine dependence – human and animal studies, current pharmacotherapies and future

perspectives

Magdalena Zaniewska, Edmund Przegaliñski, Ma³gorzata Filip

Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smêtna 12, PL 31-343 Kraków, Poland

Correspondence:Magdalena Zaniewska, e-mail: zaniew@if-pan.krakow.pl

Abstract:

Nicotine dependence is a disease of constantly growing importance. This mini-review describes the effects of nicotine in humans and focuses on the various laboratory animal models developed to study the dependence-related behavioral effects of nicotine. In ad- dition, we outline the current therapeutic approaches designed to substitute nicotine from cigarette smoke with safer compounds or to relieve symptoms of nicotine withdrawal during smoking abstinence. Since several pharmacotherapies have failed to be effica- cious in all patients, we also assess the clinical effectiveness of newer agents in relation to existing drugs.

Key words:

nicotine, drug discrimination, self-administration, reinstatement of seeking behavior, sensitization, rat, clinical trials, nicotine pharmacotherapy

Abbreviations: ACh – acetylcholine, DA – dopamine, EMEA – European Medicines Agency, FDA – Food and Drug Admin- istration, GABA –g-aminobutyric acid, Glu – glutamate, 5-HT – serotonin, IARC – International Agency for Research on Cancer, NA – noradrenaline, nACh – nicotinic acetylcholine, NRT – nicotine replacement therapy

Introduction

Addiction is a chronic disease of the central nervous system, characterized by loss of control over taking addictive substances that leads to compulsive drug- seeking and drug-taking behaviors. The disease is characterized by relapses that can be triggered even

after a very long period of abstinence and is accompa- nied by psychic, somatic and vegetative disturbances.

Addiction is associated with the distortion of the reward system, an important functional system of the brain that is physiologically responsible for feeding, water intake, and sexual and aggressive behaviors [42]. The neuroanatomical localization of the reward system is the ventral tegmental area, which contains dopaminergic neurons that send projections to the nu- cleus accumbens, amygdala and other limbic struc- tures of the brain forming the mesolimbic pathway, and to the prefrontal cortex via the dopamine (DA) mesocortical pathway [3, 42].

The reward system can be stimulated by different chemical substances that produce feelings of pleasure and euphoria and, when taken frequently, lead to ad-

Pharmacological Reports 2009, 61, 957–965 ISSN 1734-1140

Copyright © 2009 by Institute of Pharmacology Polish Academy of Sciences

(2)

Nicotine

Nicotine is an alkaloid found in the leaves of the to- bacco plant (Nicotiana); cigarettes are manufactured from the two species N. tabacum and N. rustica. The first indications of tobacco use by humans (smoking of dried tobacco leaves in a clay pipe called a tobaga by American Indian tribes) were discovered in the 15th century, after the landing of Columbus and his crew in the Caribbean. In the 16th century, tobacco was already known in the Iberian Peninsula, and its further spread through Europe was due to a French physician and diplomat, Jean Nicot de Villemain, who recommended the use of nicotine as medication.

Trace amounts of nicotine are found also in potatoes, tomatoes and sweet pepper [1]. Nicotine content in to- bacco leaves approximates 1.5 % of their dry weight.

A cigarette contains 8.4 mg of nicotine on average, of which 1–3 mg is absorbed in humans through the in- halation of the smoke [64, 74].

Nicotine was isolated from tobacco in 1828; its structure was established 15 years later, and it was synthesized in 1904 [1].

Naturally occurring nicotine is an optically active compound. Tobacco contains mostly the (S) enantio- mer ((-)-nicotine), which has a stronger biological ef- fect and is metabolized more slowly in the organism than the (R) enantiomer ((+)-nicotine) [64] (Fig. 1).

Nicotine present in tobacco smoke is inhaled to the lungs, where it is absorbed into the circulation and is

delivered to the central nervous system and other tis- sues within a few seconds. Nicotine can also be ab- sorbed in the digestive tract, mainly in the small intes- tine, as a result of swallowing saliva, and also from the skin surface [64, 74]. The highest nicotine concen- trations have been found in the brain, lungs and spleen [64, 74]. The half life of nicotine in the blood is about 120 min in humans, 55 min in rats and 10 min in mice [74, 77]. Nicotine biotransformation occurs principally in the liver; much less is metabolized in the lungs and brain [64, 74]. The main metabolite, co- tinine, is formed in the reaction catalyzed by an en- zyme CYP2A6 belonging to the cytochrome P450 family. Other metabolites include nicotine N’-oxide, trans-3’-hydroxycotinine, 5’-hydroxycotinine, cotinine N-oxide, 3-pyridyloacetic acid and nornicotine. A small fraction (5–10%) of nicotine is excreted by the kid- neys in unmetabolized form [64, 74].

Apart from nicotine, tobacco smoke contains other substances (> 4,000 compounds) (Tab. 1). They include the following compounds: crotonaldehyde, benzo(a) pyrene, formaldehyde, cresol, methanol, naphthylamine,

Fig. 1. Structural formula of nicotine ((-)-1-methyl-2-(3-pyridyl) pyrrolidine; CH"N)

Tab. 1.Composition of cigarette smoke

Components of cigarette smoke

Gaseous compounds Solid compounds

– carbon monoxide and dioxide – volatile N-nitrosamines

– nitriles and other nitrogen compounds – volatile hydrocarbons, alcohols, ketones – pyridine alkaloids (e.g., nicotine, nornicotine) – amines

– organic bases

– pyridine alkaloids (e.g., nicotine, nornicotine, anabasine)

– polycyclic aromatic hydrocarbons: e.g., pyrene, benzo(a)pyrene, benzoanthracene – metal ions and radioactive compounds (e.g., potassium, calcium, selenium, cadmium,

polonium, arsenic) – phenols – organic acids – N-nitrosamines

(3)

dimethylnitrosamine, toluene, carbon monoxide and dioxide; all of them have been classified by the Inter- national Agency for Research on Cancer (IARC) as toxic and/or carcinogenic substances. There are also free radicals, harmful radioactive compounds and heavy metals in the cigarette smoke. It is worth men- tioning that besides nicotine, tobacco smoke contains also other pharmacologically active alkaloids like nor- nicotine, anabasine, nicotirine, anatabine, cotinine and harmane [64, 73, 74]. Data from the literature indicate that the above compounds enhance the functional ef- fects of nicotine and increase the risk of addiction [74].

Smoking cigarettes causes, apart from strong ad- diction, a number of cancers (affecting lung, esopha- gus, mouth, kidney, stomach, pancreas, bone marrow) and diseases of the respiratory (chronic obstructive lung disease, emphysema, asthma, respiratory tract in- fections), digestive (stomach and duodenal ulcer) and cardiovascular (ischemic heart disease, atherosclero- sis, heart attack, cerebral stroke) systems [74].

Despite the mutagenic and carcinogenic properties of cigarette smoke, the number of cigarette smokers in Poland has been estimated at 10 million people who smoke 90 million cigarettes a day [cf. 7].

Nicotine effects in humans

In humans, nicotine affects the peripheral and central nerv- ous systems; its effects are strictly dose-dependent [74].

Peripheral effects of nicotine are attributed to the stimulation (low doses) or inhibition (high doses) of autonomic system ganglia (sympathetic and parasym- pathetic) and neuromuscular fibers. The most charac- teristic effects of lower doses of nicotine include an increase in blood pressure, tachypnea, gastric hyper- secretion or vasoconstriction. The use of higher doses induces the opposite effects, mostly hypoperistalsis and hypotension [74].

In the central nervous system, low doses of nico- tine produce neuronal stimulation, thereby increasing vigilance, learning abilities and memory performance, while the higher doses cause feelings of pleasure, dis- torted perception, photophobia, tiredness, and deficits in concentration and memory [39, 48, 62].

Extremely high nicotine doses can lead to acute in- toxication, manifested by paleness, sweating, nausea, diarrhea, headache, dizziness, muscular tremor and

confusion. In severe cases, loss of consciousness, convulsions and even death, due to respiratory center depression, may follow [58]. The lethal nicotine dose in non-smokers is 50–100 mg, while in smokers it is higher because of the development of tolerance.

Numerous studies have demonstrated the strong addictive liability of nicotine, a major component in tobacco [53]. Nicotine addiction is a complex process which in humans is comprised of the development of addiction, nicotine craving and relapses that can occur even after a very long abstinence period [44] (Tab. 2).

The development of addiction is closely associated with the rewarding and subjective effects of nicotine, while relaxation, decreased appetite, alleviation of stress and weariness symptoms, and mood elevation are other reasons why a smoker would want to have another cigarette. Social pressure (smokers’ company) and genetic factors are also important [33, 39, 44, 51, 62, 70]. It is worth emphasizing that even a single small dose of nicotine (smoking one cigarette) can in- duce a desire for the next cigarette, which quickly leads to addiction [25]. Notably, young smokers are the most vulnerable to quick development of addiction.

One of the crucial elements of nicotine addiction is development of tolerance, which forces a chronic smoker to smoke cigarettes more frequently. Such peo- ple also exhibit tolerance to the side effects of nicotine (nausea, vomiting, dizziness and headache) that are ob- served after a single nicotine use in naive smokers [74].

Abrupt nicotine withdrawal induces aversive absti- nence symptoms. A smoker desperately striving to re- lieve these negative symptoms smokes another ciga- rette [51]. The withdrawal symptoms appear within the first several hours after smoking the last cigarette and in many smokers last up to 6 months [35, 51].

They include somatic (physical) and affective signs (Tab. 2). The most frequent somatic symptoms are bradycardia and increased appetite, while the most common affective symptoms are nicotine craving, de- pressed mood, dysphoria, irritability, anxiety, and dif- ficulties concentrating [4, 41]. It is worth noting that in nicotine dependence, smoking-associated environ- mental cues (e.g., cigarette commercials or seeing other people smoking) are a significant factor contrib- uting to the increased risk of relapse.

Epidemiological data indicate that smoking to- bacco is very characteristic of poly-drug users also us- ing alcohol or cocaine, as well as in patients with schizophrenia, depression or attention deficit hyperac- tivity disorder [48, 57, 64].

Nicotine in human and animal research

Magdalena Zaniewska et al.

(4)

Besides the above-mentioned harmful effects of nicotine, there are some clinical reports suggesting that this alkaloid shows a certain neuroprotective ef- fect in Parkinson’s and Alzheimer’s disease, has therapeutic action in Huntington’s disease and Touret- te’s syndrome, reduces attention deficit in Alzheimer’s disease patients and schizophrenics, and also allevi- ates emotional disturbances and decreases pain per- ception [48, 62, 64].

Behavioral effects of nicotine in animals

A single nicotine dose produces changes in animal be- havior and other effects resulting from the activation of specific receptors. In particular, nicotine increases locomotor activity in rodents [15, 75, 78]. However, it is worth emphasizing that the locomotor effects of nicotine are dependent on the dose (low doses pro- duce a biphasic effect of initial hypomotility followed

tine was shown to decrease the body temperature in animals, to induce analgesic effects in the hot plate and tail flick tests and to enhance cognitive functions in rodents by improving attention, learning and mem- ory [5, 6, 22, 47, 74]. Nicotine also produces “anti- depressant-like” effects in the forced swim test (FST) in rodents [59, 65, 71] and effects characteristic of anxiolytic drugs, though in the latter case, the results are inconsistent [13, 60].

Recent data indicated that repeated nicotine treat- ment induces symptoms of addiction in animals [41, 44]. In particular, as in humans, addiction in animals has three distinguishable phases: development of ad- diction, abstinence signs and relapses (Tab. 2).

Different phases of nicotine dependence can be studied in various animal models. The rewarding ef- fects of nicotine are examined in the self-admini- stration procedure [17] and the conditioned place preference [43, 45]. Repeated nicotine treatment elic- its also subjective effects allowing the animals to dis- criminate between nicotine and its vehicle in the drug discrimination test [44, 67].

In animals, the withdrawal syndrome can be evoked by abrupt cessation of nicotine treatment or administration of an appropriate receptor antagonist after a period of nicotine self-administration or chronic nicotine passive administration (injections or osmotic minipumps) [41, 49, 51]. The somatic effects of withdrawal, e.g., forelimb tremor, head twitches, jumps and piloerection, appear immediately after the antagonist treatment [5, 41, 63]. The affective signs of nicotine withdrawal (e.g., anhedonia) in animals are investigated by analysis of the brain stimulation re- ward threshold for different brain structures in the in- tracranial self-stimulation model [40, 51]. Withdrawal from repeated nicotine treatment in animals also trig- gers “depressive-like” behaviors, manifested by an in- creased immobility time in the FST or “anxiogenic”

effects, such as shortening the time spent on the open arms in the elevated plus maze test [11].

Nicotine craving caused by the intake-associated environmental cues induces drug-seeking behavior and relapses in nicotine self-administration, place preference or conditioned locomotor activity mod- els/tests [43, 45, 78].

Relapses and nicotine-induced permanent neu- roadaptive changes in the brain are modeled by, re-

Humans Animals

Addiction development - rewarding effects (feeling of satisfaction)

- subjective effects (pleasant experiences)

Addiction development - rewarding effects

- subjective effects

Withdrawal symptoms - somatic

bradycardia, gastrointestinal disturbances, constipation, increased appetite, body weight gain, nausea, headache, muscular pain, tiredness

Withdrawal signs - somatic tremor of the whole

body/forelimbs, head twitches, wet dog shakes, jumps, gasps, escape attempts, genital grooming, scratching, yawning, paw licks, mastication/teeth chattering, piloerection, muscular spasms, muscle relaxation - affective

nicotine craving, depressed mood, anxiety, irritability, nervousness, aggression, concentration deficits, insomnia

- affective

anhedonia, “depressive-like”

behavior, nicotine craving, anxiety

Relapses induced by - nicotine

- stress or environmental cue

Relapses induced by - nicotine

- stress or environmental cue

(5)

spectively, the reinstatement of extinguished instru- mental response (pressing the lever associated with previous nicotine self-administration) of the animals to the priming dose of nicotine or nicotine-associated cue, or enhancement (sensitization) of locomotor ac- tivity in animals that were receiving repeated inter- mittent treatments of this substance [51, 56].

Mechanism of nicotine action

The mechanism of the pharmacological action of nicotine is based on the stimulation of nicotinic ace- tylcholine (nACh) receptors that are ion channel per- meable to monovalent (Na+, K+) and divalent (Ca++) cations. These receptors belong to the ionotropic re- ceptor family [77]. The nACh receptor is a pentamer composed of four types of subunits:a, b, g and d. In adults, the g subunit is replaced by the e subunit. Each subunit of the nACh receptor contains four transmem- brane domains. The subunits of the nACh receptor form different hetero- or homomeric combinations [38].

In mammals, nACh receptors are localized in the central nervous system, autonomic ganglia, at neuro- muscular junctions, on immune cells, keratinocytes, and intestinal and pulmonary mucosa [38]. Neuronal nACh receptors in mammals are formed by eight types of a (a2a7anda9a10) and three types of b (b2b4) subunits [38].

In the central nervous system, nACh receptors are localized on acetylcholine (ACh)-synthesizing neu- rons (somatodendritic receptors) and their terminals in many brain structures [28, 37, 69]. The nACh re- ceptors can also function as pre- and postsynaptic re- ceptors present on other neurons. It was shown that the stimulation of somatodendritic and presynaptic nACh heteroreceptors elevates the release of different neurotransmitters, while activation of postsynaptic nACh receptors mediates fast excitatory transmission [38, 62]. The nACh heteroreceptors are localized on the serotonin (5-HT) endings, mainly in the striatum and hypothalamus, on DA terminals in the striatum, and on endings of glutamatergic neurons in the mid- brain and of g-aminobutyric acid (GABAergic) neu- rons in the ventral tegmental area [3, 9, 37, 51, 62, 63, 68]. Moreover, these receptors are present on DA and GABA neurons in the ventral tegmental area and sub- stantia nigra [3, 9, 37, 51, 62, 63].

Nicotine exhibits variable affinity for nACh recep- tors containing subunitsa4b2,a3b2ora6b2(Tab. 3).

The studies performed so far have demonstrated that nACh receptors composed ofa4,a5,a6andb2subunits, and alsoa7homomeric pentamers, play an important role in the functional effects of nicotine [9, 28, 38, 62, 63].

Nicotine, by acting on its specific receptor, evokes changes in the cell membrane electrical potential, thus causing membrane depolarization and formation of an action potential [38, 77]. In vitro studies have shown that nicotine activates the spinal GABAergic and the hippocampal glutamatergic neurons [37]. Nicotine, administered systemically or iontophoretically, acti- vates the midbrain DA neurons and prolongs their fir- ing time [9, 62]. In addition, the modulating effects of nicotine on 5-HT neurons of the dorsal raphe nuclei have been described [54].

Acute systemic nicotine administration elevates synaptic release of many neurotransmitters in ani- mals, including ACh, glutamate (Glu), GABA and monoamines such as DA, noradrenaline (NA) and 5-HT [38, 62]. In addition, an increased release of opioid peptides, such as b-endorphin and met- enkephalin, has been reported in the rat brain and the striatum, respectively [23, 24]. Nicotine augments hormone secretion in rat blood, increasing the con- centrations of corticosterone, adrenocorticotropic hor- mone, prolactin, antidiuretic hormone (vasopressin) and growth hormone [18, 50, 52, 64]. Nicotine was also documented to enhance nerve growth factor syn-

Nicotine in human and animal research

Magdalena Zaniewska et al.

Tab. 3.Affinity of nicotine for the nACh receptor subunits

nACh receptor containing subunits

Affinity of nicotine KE(nM)

References

Humans Rodents

ab --- 3.6–12 [61]

ab" 9,900 83–112 [61]

a!b 14 47 [61]

a!b" 187 100–475 [61]

a"b 1–4.6 0.8–10 [61]

a"b" 6,690 40–74 [61]

a$b --- 3.8 [12]

a$b!b"a# --- 156 [61]

a% 2,000 130–15,000 [61]

Abbreviations: nACh – nicotinic acetylcholine; --- – not studied

(6)

Current pharmacotherapies for nicotine addiction

The search for an efficient pharmacotherapy for nico- tine addiction has lasted many years. However, the fact that addiction is a complex disease makes the task more complicated. Currently, the substitution strategy involving nicotine replacement therapy (NRT) in the form of chewing gum, patches, sublingual tablets, na- sal spray or inhaler (Nicotinell, Nicorette, Niquitin) has been approved by the US Food and Drug Admin- istration (FDA) and European Medicines Agency (EMEA) for the treatment of nicotine dependence. The NRT aims to replace nicotine delivered in cigarette smoke and is safer for the organism due to the lack of harmful substances present in cigarette smoke [30, 64].

Substitution therapy does not work satisfactorily for all cigarette smokers, so bupropion (Zyban) is of- ten used in smoking cessation therapy [26, 27]. This drug inhibits the reuptake of DA and NA [2]. By in- creasing the level of these monoamines in the synap- tic cleft, it may effectively prevent relapses of nico- tine use and cigarette smoking [36]. Double-blind studies performed on smokers in recent years have demonstrated that bupropion sustained-release pro- longs nicotine abstinence [21].

Another drug used in smoking cessation therapy is cytisine (Tabex). The mechanism of action of this plant-derived alkaloid is associated with partial acti- vation of the a4b2 nACh subtype receptors that evokes a moderate DA release, thereby decreasing nicotine craving when nicotine is absent, while at- tenuating its rewarding effects when it is present [77].

A promising strategy of nicotine dependence ther- apy utilizes varenicline (Champix), which, like cytisine, is a partiala4b2nACh subtype receptor ago- nist, but also a full a7 subtype receptor agonist [10, 26, 27, 55]. Varenicline has higher than nicotine affin- ity for thea4b2nACh subtype receptor and was shown to alleviate nicotine craving and to attenuate the feel- ings of pleasure associated with smoking [8, 76].

The so-called second choice treatment drugs in nicotine dependence pharmacotherapy include nor- triptyline (Allergon) and clonidine (Iporel), which

while the second is a hypotensive drug that functions as ana2-adrenoceptor agonist, used in smoking cessa- tion therapy to relieve excessive irritability during nicotine withdrawal.

The use of current pharmacotherapies is often lim- ited by the side effects observed in many patients. The adverse effects of NRT or cytisine are typical for acute intoxication with nicotine (e.g., headaches, gas- tric problems, nausea) or may include other ailments such as local irritation (nicotine patches, inhalers or aerosols) [8, 34]. The use of bupropion is associated with appearance of tachycardia, headaches, nausea, hypotension, dry mouth, gastrointestinal disturbances, insomnia and muscular tremor [36], while the use of nortriptyline or clonidine may cause dry mouth, sleepiness and tiredness [34, 36]. It should also be mentioned that varenicline produces many fewer side effects than other anti-nicotine therapies [8, 34].

The above-mentioned drugs, besides causing ad- verse effects limiting their use, have low clinical effi- cacies [30]. Therefore, there is still an urgent need to find new therapeutic strategies for the treatment of nicotine dependence, especially those that are devoid of adverse reactions and more effective than current options [29–31]. Intensive clinical trials of “mixed”

preparations composed of different drugs/substances af- fecting the ACh system, i.e., varenicline or meca- mylamine (a noncompetitive nACh receptor antagonist) in combination with NRT, or treatment with the above ACh compounds and bupropion (DA and NA reup- take inhibitor) are underway [19].

Early clinical and/or preclinical trials are focusing on the neurotransmitters other than ACh. The follow- ing compounds are under examination: a DA D3recep- tor antagonist – trans-N-[4-[2-(6-cyano-1,2,3,4-tetra- hydroisoquinolin-2-yl)ethyl]cyclohexyl]-4-quinoline- carboxamide (SB-277011-A), a NA reuptake inhibitor – reboxetine, GABABreceptor agonists – baclofen and (3-amino-2(S)-hydroxypropyl)methylphosphinic acid (CGP 44532), a monoaminooxidase A inhibitor – mo- clobemide, a monoaminooxidase B inhibitor – selegiline, a metabotropic Glu receptor subtype 5 (mGluR5) antagonist – 2-methyl-6-(phenylethynyl)-pyridine (MPEP), an opioid receptor antagonist – naltrexone, a corticotropin- releasing factor1 receptor antagonist – butyl-ethyl-[2,5-dimethyl- 7-(2,4,6-trimethylphenyl)-7H-pyrrolo [2,3-d]pyrimidin- 4-yl]amine (CP-154,526) and a cannabinoid1receptor

(7)

antagonist – rimonabant (Acomplia) [4, 19, 30]. Un- fortunately, the use of rimonabant for smoking cessa- tion therapy can be limited due to its adverse effects, such as depression episodes, anxiety and increased risk of suicidal attempts, which have been observed in overweight patients treated with this drug [14, 46].

Rimonabant was withdrawn from the market in many European countries [20] and, despite its efficacy in preclinical studies on nicotine dependence [16], it will not probably be considered as a potential therapeutic option in this type of addiction. On the other hand, since nicotine withdrawal is associated with an in- creased appetite and body weight gain, treatment has been proposed that combines rimonabant with NRT, e.g., nicotine patches [66].

The studies on vaccines that induce anti-nicotine antibodies and decrease the amount of nicotine reach- ing the brain (after its systemic administration) thereby attenuating its rewarding effects, are also worth mentioning [53]. The results of clinical studies on NicQb, NicVAX or TA-NIC vaccines seem to raise increasing hopes that such treatment strategy may be a successful one [53].

Conclusions

The currently available knowledge about nicotine de- pendence is still growing and is partly based on data from the laboratory animal models. The lack of effec- tiveness and adverse effects associated with the use of currently available anti-nicotinic drugs support the notion that researchers must continue to seek more ef- fective pharmacological treatments.

Acknowledgements:

This paper was supported by the statutory funds from the Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences (Kraków) and by the Foundation for Polish Science to M.Z.

References:

1. Andersson C, Wennström P, Gry J: Nicotine alkaloids in Solanaceous food plants. TemaNord, 2003, 531, 1–37.

2. Ascher JA, Cole JO, Colin JN, Feighner JP, Ferris RM, Fibiger HC, Golden RN et al.: Bupropion: a review of its

mechanism of antidepressant activity. J Clin Psychiatry, 1995, 56, 395–401.

3. Balfour DJ: Neuroplasticity within the mesoaccumbens dopamine system and its role in tobacco dependence. Curr Drug Targets CNS Neurol Disord, 2002, 1, 413–421.

4. Berrettini WH, Lerman CE: Pharmacotherapy and phar- macogenetics of nicotine dependence. Am J Psychiatry, 2005, 162, 1441–1451.

5. Bia³a G, Budzyñska B, Kruk M: Naloxone precipitates nicotine abstinence syndrome and attenuates nicotine- induced antinociception in mice. Pharmacol Rep, 2005, 57, 755–760.

6. Bia³a G, Kruk M: Influence of bupropion and calcium channel antagonists on the nicotine-induced memory- related response of mice in the elevated plus maze. Phar- macol Rep, 2009, 61, 236–244.

7. Biernat M: The truth about nicotine (Polish). Articles wiki: http://bioinfo.mol.uj.edu.pl/articles/Biernat05, 2006.

8. Cahill K, Stead LF, Lancaster T: Nicotine receptor partial agonists for smoking cessation. Cochrane Database Syst Rev, 2007, (1), CD006103.

9. 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, 4–7.

10. Carroll IF, Yokota Y, Wei M, Lee JR, Brieaddy LE, Bur- gess JP, Navarro HA et al.: Synthesis, nicotinic acetyl- choline receptor binding, and pharmacological properties of 3’-(substituted phenyl)deschloroepibatidine analogs.

Bioorg Med Chem, 2008, 16, 746–754.

11. Chae Y, Yeom M, Han JH, Park HJ, Hahm DH, Shim I, Lee HS et al.: Effect of acupuncture on anxiety-like be- havior during nicotine withdrawal and relevant mecha- nisms. Neurosci Lett, 2008, 430, 98–102.

12. Champtiaux N, Gotti C, Cordero-Erausquin M, David DJ, Przybylski C, Lena C, Clementi F et al.: Subunit composition of functional nicotinic receptors in dopa- minergic neurons investigated with knock-out mice.

J Neurosci, 2003, 23, 7820–7829.

13. Cheeta S, Irvine E, File SE: Social isolation modifies nicotine’s effects in animal tests of anxiety. Br J Pharma- col, 2001, 132, 1389–1395.

14. Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup A: Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. Lan- cet, 2007, 370, 1706–1713.

15. Clarke PB, Kumar R: The effects of nicotine on locomo- tor activity in non-tolerant and tolerant rats. Br J Phar- macol, 1983, 78, 329–337.

16. Cohen C, Kodas E, Griebel G: CBreceptor antagonists for the treatment of nicotine addiction. Pharmacol Bio- chem Behav, 2005, 81, 387–395.

17. Corrigall WA, Coen KM: Nicotine maintains robust self-administration in rats on a limited-access schedule.

Psychopharmacology, 1989, 99, 473–478.

18. Cruz FC, Delucia R, Planeta CS: Differential behavioral and neuroendocrine effects of repeated nicotine in ado- lescent and adult rats. Pharmacol Biochem Behav, 2005, 80, 411–417.

19. Cryan JF, Gasparini F, van Heeke G, Markou A: Non- nicotinic neuropharmacological strategies for nicotine

Nicotine in human and animal research

Magdalena Zaniewska et al.

(8)

plomie, 2007, 16, 28–40.

21. Dalsgareth OJ, Hansen NC, Søes-Petersen U, Evald T, Høegholm A, Barber J, Vestbo J: A multicenter, random- ized, double-blind, placebo-controlled, 6-month trial of bupropion hydrochloride sustained-release tablets as an aid to smoking cessation in hospital employees. Nicotine Tob Res, 2004, 6, 55–61.

22. Damaj MI, Welch SP, Martin BR: Nicotine-induced antinociception in mice: role of G-proteins and adenylate cyclase. Pharmacol Biochem Behav, 1994, 48, 37–42.

23. Davenport KE, Houdi AA, Van Loon GR: Nicotine pro- tects against mu-opioid receptor antagonism by beta- funaltrexamine: evidence for nicotine-induced release of endogenous opioids in brain. Neurosci Lett, 1990, 113, 40–46.

24. Dhatt RK, Gudehithlu KP, Wemlinger TA, Tejwani GA, Neff NH, Hadjiconstantinou M: Preproenkephalin mRNA and methionine-enkephalin content are increased in mouse striatum after treatment with nicotine. J Neuro- chem, 1995, 64, 1878–1883.

25. DiFranza JR, Savageau JA, Rigotti NA, Fletcher K, Ock- ene JK, McNeill AD, Coleman M et al.: Development of symptoms of tobacco dependence in youths: 30 month follow up data from the DANDY study. Tob Control, 2002, 11, 228–235.

26. EMEA: Guideline on the development of medicinal products for the treatment of nicotine dependence. 2007.

27. FDA Consum: The FDA approves new drug for smoking cessation. 2006, 40, 29.

28. Fowler CD, Arends MA, Kenny PJ: Subtypes of nico- tinic acetylcholine receptors in nicotine reward, depend- ence, and withdrawal: evidence from genetically modi- fied mice. Behav Pharmacol, 2008, 19, 461–484.

29. Garwood CL, Potts LA: Emerging pharmacotherapies for smoking cessation. Am J Health Syst Pharm, 2007, 64, 1693–1698.

30. George TP, O’Malley SS: Current pharmacological treat- ments for nicotine dependence. Trends Pharmacol Sci, 2004, 25, 42–48.

31. George TP, Weinberger AH: Monoamine oxidase inhibi- tion for tobacco pharmacotherapy. Clin Pharmacol Ther, 2008, 83, 619–621.

32. Green TA, Cain ME, Thompson M, Bardo MT: Environ- mental enrichment decreases nicotine-induced hyperac- tivity in rats. Psychopharmacology, 2003, 170, 235–241.

33. Harvey DM, Yasar S, Heishman SJ, Panlilio LV, Hen- ningfield JE, Goldberg SR: Nicotine serves as an effec- tive reinforcer of intravenous drug-taking behavior in hu- man cigarette smokers. Psychopharmacology, 2004, 175, 134–142.

34. Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML:

Pharmacotherapy for nicotine dependence. CA Cancer J Clin, 2005, 55, 281–299.

35. Hughes JR, Gust SW, Skoog K, Keenan RM, Fenwick JW: Symptoms of tobacco withdrawal. A replication and extension. Arch Gen Psychiatry, 1991, 48, 52–59.

37. Jones S, Sudweeks S, Yakel JL: Nicotinic receptors in the brain: correlating physiology with function. Trends Neurosci, 1999, 22, 555–561.

38. Kalamida D, Poulas K, Avramopoulou V, Fostieri E, La- goumintzis G, Lazaridis K, Sideri A et al.: Muscle and neuronal nicotinic acetylcholine receptors. Structure, func- tion and pathogenicity. FEBS J, 2007, 274, 3799–3845.

39. Kalman D: The subjective effects of nicotine: methodo- logical issues, a review of experimental studies, and rec- ommendations for future research. Nicotine Tob Res, 2002, 4, 25–70.

40. Kenny PJ, Markou A: Conditioned nicotine withdrawal profoundly decreases the activity of brain reward sys- tems. J Neurosci, 2005, 25, 6208–6212.

41. Kenny PJ, Markou A: Neurobiology of the nicotine with- drawal syndrome. Pharmacol Biochem Behav, 2001, 70, 531–549.

42. Kostowski W: Drug addiction: theories and hypotheses (Polish). Psychiatria, 2005, 2, 61–76.

43. Le Foll B, Goldberg SR: Control of the reinforcing ef- fects of nicotine by associated environmental stimuli in animals and humans. Trends Pharmacol Sci, 2005, 26, 287–293.

44. Le Foll B, Goldberg SR: Effects of nicotine in experi- mental animals and humans: an update on addictive properties. Handb Exp Pharmacol, 2009, 192, 335–367.

45. Le Foll B, Goldberg SR: Nicotine induces conditioned place preferences over a large range of doses in rats. Psy- chopharmacology, 2005, 178, 481–492.

46. Leite CE, Mocelin CA, Petersen GO, Leal MB, Thiesen FV: Rimonabant: an antagonist drug of the endocannabi- noid system for the treatment of obesity. Pharmacol Rep, 2009, 61, 217–224.

47. Levin ED, Kim P, Meray R: Chronic nicotine working and reference memory effects in the 16-arm radial maze:

interactions with Dagonist and antagonist drugs. Psy- chopharmacology, 1996, 127, 25–30.

48. Levin ED, McClernon FJ, Rezvani AH: Nicotinic effects on cognitive function: behavioral characterization, phar- macological specification, and anatomic localization.

Psychopharmacology, 2006, 184, 523–539.

49. Malin DH: Nicotine dependence: studies with a laboratory model. Pharmacol Biochem Behav, 2001, 70, 551–559.

50. Marano G, Ramirez A, Mori I, Ferrari AU: Sympathec- tomy inhibits the vasoactive effects of nicotine in con- scious rats. Cardiovasc Res, 1999, 42, 201–205.

51. Markou A: Neurobiology of nicotine dependence. Philos Trans R Soc Lond B Biol Sci, 2008, 363, 3159–3168.

52. Matta SG, Fu Y, Valentine JD, Sharp BM: Response of the hypothalamo-pituitary-adrenal axis to nicotine. Psy- choneuroendocrinology, 1998, 23, 103–113.

53. Maurer P, Bachmann MF: Vaccination against nicotine:

an emerging therapy for tobacco dependence. Expert Opin Investig Drugs, 2007, 16, 1775–1783.

54. Mihailescu S, Palomero-Rivero M, Meade-Huerta P, Maza-Flores A, Drucker-Colín R: Effects of nicotine and mecamylamine on rat dorsal raphe neurons. Eur J Phar- macol, 1998, 360, 31–36.

(9)

55. Mihalak KB, Carroll FI, Luetje CW: Varenicline is a par- tial agonist ata4b2 and a full agonist at a7 neuronal nicotinic receptors. Mol Pharmacol, 2006, 70, 801–805.

56. Miller DK, Wilkins LH, Bardo MT, Crooks PA, Dwoskin LP: Once weekly administration of nicotine produces long-lasting locomotor sensitization in rats via a nico- tinic receptor-mediated mechanism. Psychopharmacol- ogy, 2001, 156, 469–476.

57. Myers MG, Kelly JF: Cigarette smoking among adoles- cents with alcohol and other drug use problems. Alcohol Res Health, 2006, 29, 221–227.

58. NIDA Research Report: Nicotine addiction. 1998.

59. Nowakowska E, Kus K, Florek E, Czubak A, Jodynis- Liebert J: The influence of tobacco smoke and nicotine on antidepressant and memory-improving effects of ven- lafaxine. Hum Exp Toxicol, 2006, 25, 199–209.

60. Ouagazzal AM, Kenny PJ, File SE: Modulation of be- haviour on trials 1 and 2 in the elevated plus-maze test of anxiety after systemic and hippocampal administration of nicotine. Psychopharmacology, 1999, 144, 54–60.

61. PDSP database: http://pdsp.med.unc.edu/indexR.html.

62. Picciotto MR: Nicotine as a modulator of behavior: be- yond the inverted U. Trends Pharmacol Sci, 2003, 24, 493–499.

63. Picciotto MR, Caldarone BJ, King SL, Zachariou V:

Nicotinic receptors in the brain. Links between molecu- lar biology and behavior. Neuropsychopharmacology, 2000, 22, 451–465.

64. Pogocki D, Ruman T, Danilczuk M, Danilczuk M, Ce- luch M, Wa³ajtys-Rode E: Application of nicotine enanti- omers, derivatives and analogues in therapy of neurode- generative disorders. Eur J Pharmacol, 2007, 563, 18–39.

65. Popik P, Krawczyk M, Kos T, Nalepa I, Kowalska M, Witarski T, Antkiewicz-Michaluk L, Vetulani J: Nicotine produces antidepressant-like actions: Behavioral and neurochemical evidence. Eur J Pharmacol, 2005, 515, 128–133.

66. Rigotti NA, Gonzales D, Dale LC, Lawrence D, Chang Y, CIRRUS Study Group: A randomized controlled trial of adding the nicotine patch to rimonabant for smoking cessation: efficacy, safety and weight gain. Addiction, 2009, 104, 266–276.

67. Rosecrans JA, Villanueva HF: Discriminative stimulus properties of nicotine: mechanisms of transduction.

NIDA Res Monogr, 1991, 116, 101–116.

68. Schwartz RD, Lehmann J, Kellar KJ: Presynaptic nico- tinic cholinergic receptors labeled by [!H]acetylcholine on catecholamine and serotonin axons in brain. J Neuro- chem, 1984, 42, 1495–1498.

69. Seth P, Cheeta S, Tucci S, File SE: Nicotinic-serotonergic interactions in brain and behaviour. Pharmacol Biochem Behav, 2002, 71, 795–805.

70. Siemiñska A: Genetic factors influencing tobacco depend- ence (Polish). Alergia Astma Immunologia, 2005, 10, 69–73.

71. Suemaru K, Yasuda K, Cui R, Li B, Umeda K, Amano M, Mitsuhashi H et al.: Antidepressant-like action of nicotine in forced swimming test and brain serotonin in mice. Physiol Behav, 2006, 88, 545–549.

72. Terry AV Jr, Clarke MS: Nicotine stimulation of nerve growth factor receptor expression. Life Sci, 1994, 55, 91–98.

73. Touiki K, Rat P, Molimard R, Chait A, de Beaurepaire R:

Harmane inhibits serotonergic dorsal raphe neurons in the rat. Psychopharmacology, 2005, 182, 562–569.

74. US DHHS: The Health Consequences of Smoking: Nico- tine Addiction: A Report of the Surgeon General. 1988.

75. Vezina P, McGehee DS, Green WN: Exposure to nicotine and sensitization of nicotine-induced behaviors. Prog Neu- ropsychopharmacol Biol Psychiatry, 2007, 31, 1625–1638.

76. West R, Baker CL, Cappelleri JC, Bushmakin AG: Effect of varenicline and bupropion SR on craving, nicotine withdrawal symptoms, and rewarding effects of smoking during a quit attempt. Psychopharmacology, 2008, 197, 371–377.

77. Wonnacott S, Barik J: Nicotinic ACh receptors. Tocris Bioscience Scientific Review Series, 2007, 28, 1–18.

78. Zaniewska M, McCreary AC, Filip M: Interactions of se- rotonin (5-HT) receptor-targeting ligands and nicotine:

locomotor activity studies in rats. Synapse, 2009, 63, 653–661.

Received:

July 24, 2009; in revised form: November 23, 2009.

Nicotine in human and animal research

Magdalena Zaniewska et al.

Cytaty

Powiązane dokumenty

Searching for variants with evidence of regulatory functions, we have reported interactions between CHRNA5 and CHRNA3 enhancer variants (tagged by rs880395 and rs1948, respectively)

Wśród badanych 21% paliło tytoń, mężczyźni częściej niż kobiety palili (25% vs. 17%) i wykazywali wyższy stopień uzależnienia.. Problemy z alkoholem wykazano u co

Badania przeprowadzono przy użyciu autorskiego kwestionariusza ankiety (zawierającego między innymi pytania dotyczące danych socjodemograficznych i motywów rzucania

test Czterech „C” (Compulsion, Control, Cutting down, Consequences) [8], Kwestionariusz Toleran- cji Fagerströma (FTQ, Fagerström Tolerance Questionnaire) oraz jego skrócone

Celem badania była ocena wpływu niektórych czynników demogra- ficznych i socjalnych na stopień uzależnienia od nikotyny i motywację do rzucenia palenia tytoniu u

program terapii palaczy tytoniu w oparciu o bupropion SR – pierwszy nie zawierający nikotyny lek pierwszego rzutu w leczeniu uzależnienia od tytoniu (5,10,11).. Celem programu

In this study we aimed to determine whether a difference in complications between smok- ers and non-smokers exists in type 2 diabetes mellitus (T2DM) and to evaluate if there is

However, in combination studies rats pretreated with a separate dose of ABT-239 (1 mg/kg) prior to nicotine (0.4 mg/kg) for 5 days and then challenged with nicotine (0.4 mg/kg) after