• Ei tuloksia

The development of tolerance in nicotine-infused mice

The hypothermic effect of nicotine is mediated by central nAChRs (Mansner et al.

1974) and can be prevented by elevating the ambient temperature (Haikala et al.

1986). In several studies chronic nicotine treatment was found to induce tolerance to the nicotine-induced decrease of body temperature (Mansner et al. 1974; Marks and Collins 1985). Further, in mice withdrawn for 24 h from a seven-week oral nicotine administration schedule (Pietilä et al. 1996), acute nicotine challenge induced significantly less hypothermia than in controls indicating the development of tolerance to the hypothermic effect of nicotine. Tolerance also developed to the locomotor-depressant effects of acute nicotine in these mice (Pietilä et al. 1998). In Study IV, acute nicotine doses of 1 and 2 mg/kg induced hypothermia in a dose-dependent manner which was at its maximum at 30 min after administration of the drug. In mice infused chronically with nicotine from subcutaneously implanted nicotine-releasing reservoirs for 7 days, tolerance was found to the hypothermic effect of the acute nicotine dose of 1 mg/kg but not to the dose of 2 mg/kg. Previously no tolerance was observed to the hypothermic effect of large or repeated (3 mg/kg or 1 x 4 mg/kg s.c.) acute nicotine doses after 7-day chronic nicotine infusion in mice (Leikola-Pelho et al.

1990). Thus, we found that increasing the nicotine dose overcomes the attenuation of

the hypothermic effect of nicotine. This suggests that tolerance, in the classical sense, i.e. an effect that is characterised by the fact that increasing the dose of the drug overcomes the diminution of its effect following repeated administration, develops to nicotine-hypothermia during chronic treatment. In contrast, during chronic nicotine treatment, acute nicotine did not increase the concentrations of striatal DA metabolites suggesting that desensitization is the main mechanism by which the nAChRs regulating striatal DA metabolism, are inactivated under the present experimental conditions. Thus, the nAChRs involved in these two effects differ.

7 SUMMARY AND CONCLUSIONS

In the present study both desensitization and long-lasting inactivation of the nAChRs involved in the mediation of nicotine’s central effects on cerebral dopamine metabolism and on Fos expression were demonstrated in vivo in rodent brain during chronic nicotine administration and its withdrawal.

1) In rat acute administration of nicotine induced Fos IS both in the CPU and limbic dopaminergic areas. Acutely administered nicotine also induced Fos IS in both the PVN and SON, which are stress-related brain areas, and in MT and IPN. Diazepam induced Fos IS in all stress-related areas studied (PVN, SON, ACe), but not in any of the other brain areas studied. No significant interactions were found between the acute effects of diazepam and nicotine on Fos expression and this finding suggests that these drugs activate different sets of neurons within the stress-related brain areas.

2) The results from studies performed in the rat suggest that the degree of desensitization of nAChRs in various brain areas differs. The nAChRs in the striatal areas seem to be desensitized more easily than those in the limbic areas during prolonged nicotine infusion. Thus, in the CPU area neither the elevation of DA metabolites nor the expression of Fos protein was affected by acute nicotine during chronic nicotine infusion. In contrast, acute nicotine administration caused some elevation of both limbic HVA and Fos expression in the NAcc of rats during chronic nicotine infusion. No reduction of nicotine-induced Fos expression was found in the Cg and ACe, and only a slight reduction was found in the IPN which are known to be brain areas involved in regulation of various behavioural reactions. Further, the nAChRs in the hypothalamic areas, PVN and SON, or in their input areas regulating nicotine’s effects on hormone secretion also seem to be more easily desensitized than the nAChRs in the limbic areas.

We also found evidence for a long-lasting inactivation of nAChRs mediating nicotine’s effects in vivo. Thus, at 24 h after withdrawal acute nicotine did not elevate limbic DOPAC, but striatal DOPAC and HVA as well as limbic HVA were elevated to the same degree as in control rats. Furthermore, acute nicotine did not increase the Fos

expression in the CPU or NAcc, although in ACe its effects were similar to that in controls.

After longer withdrawal (72 h) nicotine’s effects on cerebral dopamine metabolism and Fos protein expression were restored showing that these functional changes of nAChRs are reversible phenomena.

3) In mice, the responses of striatal DA metabolism and body temperature to acute nicotine were both reduced during constant nicotine infusion. However, the tolerance to nicotine’s hypothermic effect could be overcome to some degree by increasing the dose of nicotine, suggesting that tolerance, in the classical sense, develops in nAChRs involved in thermoregulation during chronic nicotine treatment. In contrast, during chronic nicotine treatment acute nicotine did not increase the concentrations of striatal DA metabolites suggesting that desensitization is the main mechanism by which nAChRs regulating striatal DA metabolism are inactivated under the present experimental conditions. Thus, the nAChRs involved in these two effects differ.

In conclusion, there seems to be variations in the functional states and/or in the subunit combinations of nAChRs mediating nicotine’s effects in brain. It has been hypothesized that smokers, seeking the most rewarding effect, adjust their way of smoking so that they reach the appropriate combination of stimulated and desensitized nAChRs. The long-lasting inactivation of nAChRs is one of the important factors in the development of tolerance to nicotine. The variations in responses to nicotine may prove to be useful when studying the protective effect of nicotine in Parkinson’s disease as well as in its reinforcing and dependence-producing properties.

ACKNOWLEDGEMENTS

This work was carried out at the Division of Pharmacology and Toxicology, Department of Pharmacy, University of Helsinki.

I express my deepest gratitude to my tutor and supervisor, Professor Liisa Ahtee, for her inspiring enthusiasm and encouragement. Her profound knowledge of neuropharmacology combined with great teaching skills have made the completion of this work possible. I will always value the hospitality shown to me by Prof. Ahtee whilst preparing papers to be published.

Docent Jouni Sirviö and docent Sampsa Vanhatalo offered constructive criticism on the manuscript and valuable suggestions which led to its improvement, and to them I extend my sincere thanks. I am furthermore grateful to Mr. Adrian Mogg (University of Bath, United Kingdom) for revising the language of the manuscript.

I am deeply grateful to the co-authors of these studies, M. Sc. Tiina Seppä, M. Sc.

Helena Gäddnäs and M. Sc. Sirpa Lahtinen, for their excellent collaboration and fruitful discussions during the study. To Ph. D. Kirsi Pietilä I want to extend my sincere thanks for first being my tutor and then for excellent collaboration in the field of nicotine research. To M. Sc. Mikaela Moed I express my appreciation for her capable assistance during the course of her pro gradu study. Further, I wish to thank Ms. Marjo Vaha for her skilful assistance in all stages of this work.

I was introduced to the world of Fos immunohistochemistry during a stay at the University of Cambridge, where I visited the Department of Anatomy, the laboratory of Dr. Joe Herbert in spring 1992. I am deeply grateful to him and his skilful staff for guidance in my first steps in the field of immunohistochemistry. Further thanks go to Prof. Pertti Panula and his staff at the Department of Anatomy, University of Helsinki at that time to help me with setting up the method. I wish to thank Ph. Lic. Juhani Tuominen for his advice concerning statistical problems.

Special thanks go to M. Sc. Ulla-Mari Parkkisenniemi for great friendship both in the work and leisure. I wish also to thank the entire staff of the Department of Pharmacology and Toxicology for creating a friendly and inspiring atmosphere in which it has been pleasant to work.

Finally, I owe my warmest gratitude to my husband Arto and to our children Laura and Riikka for their encouragement, support and never-ending love during the many years of the research. They certainly have kept my feet firmly on the ground. Also, the support, encouragement and love of my dear parents Raili and Jussi and my dear brother Timo has been essential. Further, I want to express sincere thanks to Minna Nieminen for being invaluable friend and support during all these years.

Financial support from University of Helsinki, the Finnish Cultural Foundation and the Research Council for Health of the Academy of Finland is gratefully acknowledged.

Helsinki, July 2000

Outi Salminen

REFERENCES

Adler LE, Olincy A, Waldo M, Harris JG, Griffith J, Stevens K, Flach K, Nagamoto H, Bickford P, Leonard S and Freedman R (1998) Schizophrenia, sensory gating and nicotinic receptors.

Schizophrenia Bull 24: 189-202.

Agid Y, Javoy F and Youdim MBH (1973) Monoamine oxidase and aldehyde dehydrogenase activity in the striatum of rats after 6-hydroxydopamine lesion of the nigrostriatal pathway. Br J Pharmacol 48: 175-178.

Akaike A, Tamura Y, Yokota T, Shimohama S and Kimura J (1994) Nicotine-induced protection of cultured cortical neurons against N-methyl-D-aspartate receptor-mediated glutamate cytotoxicity. Brain Res 644: 181-187.

Alkondon M, Pereira EFR, Barbosa CTF and Albuquerque EX (1997) Neuronal nicotinic acetylcholine receptor activation modulates gamma-aminobutyric acid release from CA1 neurons of rat hippocampal slices. J Pharmacol Exp Ther 283: 1396-1411.

Amstrong ME (1985). Cholinergic innervation of vasopressin-containing neurons in the supraoptic nucleus of the rat. In Schrier. RW (Ed.), Vasopressin, pp. 353-360. Raven Press, New York.

Andersson K, Fuxe K and Agnati LF (1981a) Effects of single injections of nicotine on the ascending dopamine pathways in the rat. Evidence for increases of dopamine turnover in the mesostriatal and mesolimbic dopamine neurons. Acta Physiol Scand 112: 345-347.

Andersson K, Fuxe K, Agnati LF and Eneroth P (1981b) Effects of acute central and peripheral administration of nicotine on ascending dopamine pathways in the male rat brain. Evidence for nicotine induced increases of dopamine turnover in various telencephalic dopamine nerve terminal systems. Med Biol 59: 170-6.

Andersson K, Siegel R, Fuxe K and Eneroth P (1983) Intravenous injections of nicotine induced very rapid and discrete reductions of hypothalamic catecholamine levels associated with increases of ACTH, vasopressin and prolactin secretion. Acta Physiol Scand 118: 35-40.

Armstrong DM, Saper CB, Levey AI, Wainer BH and Terry RD (1983) Distribution of cholinergic neurons in rat brain: demonstrated by the immunocytochemical localization of choline acetyltransferase. J Comp Neurol 216: 53-68.

Arqueros L, Naquira D and Zunino E (1978) Nicotine-induced release of catecholamines from rat hippocampus and striatum. Biochem Pharmacol 27: 2667-2674.

Balfour DJ (1991) The influence of stress on psychopharmacological responses to nicotine. Br J Addict 86: 489-93.

Balfour DJ (1994) Neural mechanisms underlying nicotine dependence. Addiction 89: 1419-23.

Balfour DJ, Benwell ME, Birrell CE, Kelly RJ and Al-Aloul M (1998) Sensitization of the mesoaccumbens dopamine response to nicotine. Pharmacol Biochem Behav 59: 1021-30.

Balfour DJK (1989) Influence of nicotine on the release of monoamines in the brain. Prog in Brain Res 79:

165-172.

Balfour DJK, Khullar AK and Longden A (1975) Effects of nicotine on plasma corticosterone and brain amines in stressed and unstressed rats. Pharmacol Biochem Behav 3: 179-184.

Beck CH and Fibiger HC (1995) Conditioned fear-induced changes in behavior and in the expression of the immediate early gene c-fos: with and without diazepam pretreatment. J Neurosci 15: 709-720.

Benowitz NL (1990). Pharmacokinetic considerations in understanding nicotine dependence. In Marsh J and Bock G (Eds.), Biology of Nicotine Dependence, Vol. 152, pp. 186-200. John Wiley, New York.

Benowitz NL (1996) Pharmacology of nicotine: Addiction and therapeutics. Annu Rev Pharmacol Toxicol 36:

597-613.

Benwell ME and Balfour DJ (1992) The effects of acute and repeated nicotine treatment on nucleus accumbens dopamine and locomotor activity. Br J Pharmacol 105: 849-56.

Benwell ME and Balfour DJ (1997) Regional variation in the effects of nicotine on catecholamine overflow in rat brain. Eur J Pharmacol 325: 13-20.

Benwell ME, Balfour DJ and Birrell CE (1995) Desensitization of the nicotine-induced mesolimbic dopamine responses during constant infusion with nicotine. Br J Pharmacol 114: 454-60.

Benwell ME, Balfour DJ and Khadra LF (1994) Studies on the influence of nicotine infusions on mesolimbic dopamine and locomotor responses to nicotine. Clin Investig 72: 233-9.

Benwell MEM and Balfour DJK (1979) Effects of nicotine administration and its withdrawal on plasma corticosterone and brain 5-hydroxyindoles. Psychopharmacology (Berl) 63: 7-11.

Benwell MEM, Balfour DJK and Anderson JM (1988) Evidence that tobacco smoking increases the density of (-)-3H-nicotine binding sites in human brain. J Neurochem 50: 1243-1247.

Bernard V, Dumartin B, Lamy E and Bloch B (1993) Fos immunoreactivity after stimulation of inhibition of muscarinic receptors indicates anatomical specificity for cholinergic control of striatal efferent neurons and cortical neurons in the rat. Eur J Neurosci 5: 1218-1225.

Berretta S, Robertson HA and Graybiel AM (1992) Dopamine and glutamate agonists stimulate neuron-spesific expression of Fos-like protein in the striatum. J Neurophysiol 68: 767-777.

Bertrand D, Galzi JL, Devillers-Thiery A, Bertrand S and Changeux J-P (1993) Mutations at two distinct sites within the channel domain M2 alter calcium permeability of neuronal α7 nicotinic receptor. Proc Natl Acad Sci 90: 6971-6975.

Binn KE (1999) The synaptic pharmacology underlying sensory processing in the superior colliculus. Prog in Neurobiol 59: 129-159.

Björklund A and Lindvall O (1984). Dopamine-containing systems in the CNS. In A Björklund and T Hökfelt (Eds.), Classical transmitters in the CNS, Part I, Vol. 2, pp. 55-122. Elsevier Science Publishers, London..

Boyd RT (1997) The molecular biology of neuronal nicotinic acetylcholine receptors. Crit Rev Toxicol 27: 299-318.

Brazell MP, Mitchell SN, Joseph MH and Gray JA (1990) 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 29: 1177-85.

Breese CR, Marks MJ, Logel J, Adams CE, Sullivan B, Collins AC and Leonard S (1997) Effect of smoking history on [3H]nicotine binding in human postmortem brain. J Pharmacol Exp Ther 282: 7-13.

Brioni JD, Kim DJ, Brodie MS, Decker MW and Arneric SP (1995) ABT-418: discriminative stimulus properties and effect on ventral tegmental cell activity. Psychopharmacology (Berl) 119: 368-75.

Brodie M (1991). Low concentration of nicotine increase the firing rate of neurons of the rat ventral tegmental area in vitro. In F Adlkofer and K Thurau (Eds.), Effects of nicotine on biological systems., pp. 373-377.

Birkhäuser, Basel.

Brown DA, Docherty RJ and Haliwell JV (1984) The action of cholinomimetic substances on impulse conduction in the habenulo-interpeduncular pathway of the rat in vitro. J Physiol (Lond) 353: 101-109.

Brown MR and Gray TS (1988) Peptide injections into the amygdala of conscious rats: effects on blood pressure, heart rate and plasma catecholamines. Regul Peptides 21: 95-106.

Bullitt E (1990) Expression of C-fos-like protein as a marker for neuronal activity following noxious stimulation in the rat. J Comp Neurol 296: 517-530.

Butcher LJ (1995). Cholinergic neurons and networks. In G Paxinos (Ed.), The rat nervous system., pp. 1003-1015. Academic Press, London.

Cachelin AB and Colquhoun D (1989) Desensitization of the acetylcholine receptor of frog end-plates measured in a vaseline-gap voltage clamp. J Physiol 415: 159-188.

Cachelin AB and Jaggi R (1991) β subunits determine the time course of desensitization in rat alpha3 neuronal nicotinic acetylcholine receptors. Pflügers Arch 419: 579-582.

Cadoni C and Di Chiara G (2000) Differential changes in accumbens shell and core dopamine in behavioral sensitizatioin to nicotine. Eur J Pharmacol 387: R23-R25.

Calabresi P, Lacey MG and North RA (1989) Nicotinic excitation of rat ventral tegmental neurones in vitro studied by intracellular recording. Br J Pharmacol 98: 135-40.

Canzek V, Wolfensberger M, Amsler U and Cuenod M (1981) In vivo release of glutamate and aspartate following optic nerve stimulation. Nature 293: 572-574.

Changeux J-P, Devillers-Thiery A and Chemouilli P (1984) Acetylcholine receptor: an allosteric protein.

Science 225: 1335-1345.

Changeux J-P, Giraudat J and Dennis M (1987) The nicotinic acetylcholine receptor: molecular architecture of a ligand-regulated ion channel. Trends in Pharmacol Sci 8: 459-465.

Chaudhuri A (1997) Neural activity mapping with inducible transcription factors. Neuroreport 8: iii-vii.

Clarke PB, Hommer DW, Pert A and Skirboll LR (1985a) Electrophysiological actions of nicotine on substantia nigra single units. Br J Pharmacol 85: 827-35.

Clarke PB and Reuben M (1996) Release of [3H]-noradrenaline from rat hippocampal synaptosomes by nicotine: mediation by different nicotinic receptor subtypes from striatal [3H]-dopamine release. Br J Pharmacol 117: 595-606.

Clarke PBS (1990) Dopaminergic mechanisms in the locomotor stimulant effects of nicotine. Biochem Pharmacol 40: 1427-1432.

Clarke PBS (1993) Nicotinic receptors in mammalian brain: localization and relation to cholinergic innervation.

Prog in Brain Res 98: 77-83.

Clarke PBS and Kumar R (1983) The effects of nicotine on locomotor activity in non-tolerant and tolerant rats. Br J Pharmacol 78: 329-337.

Clarke PBS, Schwartz RD, Paul SM, Pert CB and Pert A (1985b) Nicotinic binding in rat brain:

autoradiographic comparison of 3H-acetylcholine, 3H-nicotine and 125I-α-bungarotoxin. J Neurosci 5:

1307-1315.

Collins AC, Bhat RV, Pauly JR and Marks MJ (1990a). Modulation of nicotine receptors by chronic exposure to nicotinic agonists and antagonists, The Biology of Nicotine Dependence, Vol. 152, pp. 68-82. Wiley, New York.

Collins AC, Luo Y, Selvaag S and Marks MJ (1994) Sensitivity to nicotine and brain nicotinic receptors are altered by chronic nicotine and mecamylamine infusion. J Pharmacol Exp Ther 271: 125-133.

Collins AC and Marks MJ (1987). The effects of chronic nicotine administration on brain nicotinic receptor numbers. In WR Martin, GR Van Loon, ET Iwamoto and L Davis (Eds.), Tobacco smoking and nicotine, pp. 439-450. Plenum Publishing Corporation, New York.

Collins AC and Marks MJ (1996) Are nicotinic receptors activated or inhibited following chronic nicotine treatment? Drug Dev Res 38: 231-242.

Collins AC, Miner LL and Marks MJ (1988a) Genetic influences on acute responses to nicotine and nicotine tolerance in the mouse. Pharmacol Biochem Behav 30: 269-78.

Collins AC, Romm E and Wehner JM (1988b) Nicotine tolerance: an analysis of the time course of its development and loss in the rat. Psychopharmacology (Berl) 96: 7-14.

Collins AC, Romm E and Wehner JM (1990b) Dissociation of the apparent relationship between nicotine tolerance and up-regulation of nicotinic receptor. Brain Res Bull 25: 373-379.

Colquhoun LM and Patrick JW (1997) Pharmacology of neuronal nicotinic acetylcholine receptor subtypes.

Adv Pharmacol 39: 191-220.

Conte-Devolx B, Oliver C, Giraud P, Gillioz P, Castanas E, Lissitzky JC, Bourdouresque F and Millet Y (1981) Effect of nicotine on in vivo secretion of melanocorticotropic hormones in the rat. Life Sci 28:

1067-1073.

Contestabile A and Flumerfelt BA (1981) Afferent connections of the interpeduncular nucleus and the topographic organization of the habenulo-interpeduncular pathway: an HRP study in the rat. J Comp Neurol 196: 253-270.

Cooper JR, F.E. B and Roth RH (1996) The biochemical basis of neuropharmacology, 7th ed. Oxford University Press, New York.

Couturier S, Bertrand D, Matter J-M, Hernandez M-C, Bertrand S, Millar N, Valera S, Barkas T and Ballivet M (1990) A neuronal nicotinic acetylcholine receptor subunit (α7) is developmentally regulated and forms a homo-oligomeric channel blocked by alpha-BTX. Neuron 5: 847-856.

Cox BM (1990). Drug tolerance and physical dependence. In W Pratt and P Taylor (Eds.), Principles of Drug Action, pp. 639-690. Churchill Livingstone, New York.

Curran T and Franza BR (1988) Fos and Jun: the AP-1 connection. Cell 55: 395-397.

Curran T, MacConnell WP, van Straaten F and Verma IM (1983) Structure of the FBJ murine osteosarcoma virus genome: Molecular cloning of its associated helper virus and the cellular homolog of the v-fos gene from mouse and human cells. Mol Cell Biol 3: 914-921.

Curran T and Morgan JI (1987) Memories of fos. BioEssays 7: 255-258.

Curvall M, Kazemi-Vala E and Enzell CR (1982) Simultaneous determination of nicotine and cotinine in plasma using capillary column gas chromatography with nitrogen-sensitive detector. J Chromatogr 232:

283-293.

Dahlström A and Fuxe K (1964) Evidence for the existence of monoamine containing neurons in the central nervous system. 1. Demonstration of monoamines in the cell bodis of brain stem neurons. Acta Physiol Scand 62: 1-55.

Dalack GW, Healy DJ and Meador-Woodruff JH (1998) Nicotine dependence in schizophrenia: clinical phenomena and laboratory findings. Am J Psychiatry 155: 1490-501.

Damsma G, Day J and Fibiger HC (1989) Lack of tolerance to nicotine-induced dopamine release in the nucleus accumbens. Eur J Pharmacol 168: 363-8.

Damsma G, Westerink BH, de Vries JB and Horn AS (1988) The effect of systemically applied cholinergic drugs on the striatal release of dopamine and its metabolites, as determined by automated brain dialysis in conscious rats. Neurosci Lett 89: 349-54.

Dani JA and Heinemann S (1996) Molecular and cellular aspects of nicotine abuse. Neuron 16: 905-908.

Decker MW, Brioni JD, Bannon AW and Arneric SP (1995) Diversity of neuronal nicotinic acetylcholine

Decker MW, Brioni JD, Bannon AW and Arneric SP (1995) Diversity of neuronal nicotinic acetylcholine