A. Agmo and M. Gomez, Conditioned place preference produced by infusion of Met-enkephalin into the medial preoptic area, Brain Res, vol.550, pp.343-346, 1991.

A. Agmo and M. Gomez, Sexual reinforcement is blocked by infusion of naloxone into the medial preoptic area, Behav Neurosci, vol.107, pp.812-818, 1993.

S. H. Ahmed, P. J. Kenny, G. F. Koob, and A. Markou, Neurobiological evidence for hedonic allostasis associated with escalating cocaine use, Nat Neurosci, vol.5, pp.625-626, 2002.

S. H. Ahmed and G. F. Koob, Transition from moderate to excessive drug intake: change in hedonic set point, Science, vol.282, pp.298-300, 1998.

H. Akil, C. Owens, H. Gutstein, L. Taylor, E. Curran et al., Endogenous opioids: overview and current issues, Drug Alcohol Depend, vol.51, pp.127-140, 1998.

G. F. Alheid and L. Heimer, New perspectives in basal forebrain organization of special relevance for neuropsychiatric disorders: the striatopallidal, amygdaloid, and corticopetal components of substantia innominata, Neuroscience, vol.27, pp.1-39, 1988.

A. Fischer, D. Schafer, M. K. Ferger, B. Gross, S. Westermann et al., Sensitization to the behavioural effects of cocaine: alterations in tyrosine hydroxylase or endogenous opioid mRNAs are not necessarily involved, Naunyn-Schmiedebergs Arch Pharmacol, vol.363, pp.288-294, 2001.

S. Ammon-treiber and V. Hollt, Morphine-induced changes of gene expression in the brain, Addict Biol, vol.10, pp.81-89, 2005.

A. Argiolas, Neuropeptides and sexual behaviour, Neurosci Biobehav Rev, vol.23, pp.1127-1142, 1999.

A. Argiolas and M. R. Melis, Central control of penile erection: role of the paraventricular nucleus of the hypothalamus, Prog Neurobiol, vol.76, pp.1-21, 2005.

M. Arroyo, W. A. Baker, and B. J. Everitt, Cocaine self-administration in rats differentially alters mRNA levels of the monoamine transporters and striatal neuropeptides, Brain Res, vol.83, pp.107-120, 2000.

M. Arroyo, A. Markou, T. W. Robbins, and B. J. Everitt, Acquisition, maintenance and reinstatement of intravenous cocaine self-administration under a second-order schedule of reinforcement in rats: effects of conditioned cues and continuous access to cocaine, Psychopharmacology, vol.140, pp.331-344, 1998.

, Association AP. Diagnostic and Statistical Manual of Mental Disorders. 4, 2000.

G. Aston-jones, J. M. Delfs, J. Druhan, and Y. Zhu, The bed nucleus of the stria terminalis. A target site for noradrenergic actions in opiate withdrawal, Ann NY Acad Sci, vol.877, pp.486-498, 1999.

M. R. Azar, B. C. Jones, and G. Schulteis, Conditioned place aversion is a highly sensitive index of acute opioid dependence and withdrawal, Psychopharmacology, vol.170, pp.42-50, 2003.

A. V. Azaryan, B. J. Clock, J. G. Rosenberger, and B. M. Cox, Transient upregulation of mu opioid receptor mRNA levels in nucleus accumbens during chronic cocaine administration, Can J Physiol Pharmacol, vol.76, pp.278-283, 1998.

A. V. Azaryan, L. J. Coughlin, B. Buzas, B. J. Clock, and B. M. Cox, Effect of chronic cocaine treatment on mu-and delta-opioid receptor mRNA levels in dopaminergically innervated brain regions, J Neurochem, vol.66, pp.443-448, 1996.

A. Bailey, V. Yuferov, J. Bendor, S. D. Schlussman, Y. Zhou et al., Immediate withdrawal from chronic "binge" cocaine administration increases mu-opioid receptor mRNA levels in rat frontal cortex, Brain Res, vol.137, pp.258-262, 2005.

B. A. Baldo and A. E. Kelley, Discrete neurochemical coding of distinguishable motivational processes: insights from nucleus accumbens control of feeding, Psychopharmacology, vol.191, pp.439-459, 2007.

B. W. Balleine, Neural bases of food-seeking: affect, arousal and reward in corticostriatolimbic circuits, Physiol Behav, vol.86, pp.717-730, 2005.

B. W. Balleine and S. Killcross, Parallel incentive processing: an integrated view of amygdala function, Trends Neurosci, vol.29, pp.272-279, 2006.

R. Bals-kubik, A. Ableitner, A. Herz, and T. S. Shippenberg, Neuro-anatomical sites mediating the motivational effects of opioids as mapped by the conditioned place preference paradigm in rats, J Pharmacol Exp Ther, vol.264, pp.489-495, 1993.

J. Balthazart and G. F. Ball, Topography in the preoptic region: differential regulation of appetitive and consummatory male sexual behaviors, Front Neuroendocrinol, vol.28, pp.161-178, 2007.

M. F. Barbano and M. Cador, Opioids for hedonic experience and dopamine to get ready for it, Psychopharmacology, vol.191, pp.497-506, 2007.

M. T. Bardo, Neuropharmacological mechanisms of drug reward: beyond dopamine in the nucleus accumbens, Crit Rev Neurobiol, vol.12, pp.37-67, 1998.

R. Basheer and A. Tempel, Morphine-induced reciprocal alterations in G alpha s and opioid peptide mRNA levels in discrete brain regions, J Neurosci Res, vol.36, pp.551-557, 1993.

A. S. Beadles-bohling and K. M. Wiren, Alteration of kappa-opioid receptor system expression in distinct brain regions of a genetic model of enhanced ethanol withdrawal severity, Brain Res, vol.1046, pp.77-89, 2005.

J. Beaulieu, D. Champagne, and G. Drolet, Enkephalin innervation of the paraventricular nucleus of the hypothalamus: distribution of fibers and origins of input, J Chem Neuroanat, vol.10, pp.79-92, 1996.

D. M. Bronstein, R. Przewlocki, and H. Akil, Effects of morphine treatment on pro-opiomelanocortin systems in rat brain, Brain Res, vol.519, pp.102-111, 1990.

I. Broseta, M. Rodriguez-arias, L. Stinus, and J. Minarro, Ethological analysis of morphine withdrawal with different dependence programs in male mice, Prog Neuropsychopharmacol Biol Psychiatry, vol.26, pp.335-347, 2002.

B. Buzas, J. Rosenberger, and B. M. Cox, Mu and delta opioid receptor gene expression after chronic treatment with opioid agonist, Neuroreport, vol.7, pp.1505-1508, 1996.

E. M. Byrnes, Chronic morphine exposure during puberty induces long-lasting changes in opioidrelated mRNA expression in the mediobasal hypothalamus, Brain Res, vol.1190, pp.186-192, 2008.

A. Cabral, N. Isoardi, C. Salum, C. E. Macedo, M. J. Nobre et al., Fear state induced by ethanol withdrawal may be due to the sensitization of the neural substrates of aversion in the dPAG, Exp Neurol, vol.200, pp.200-208, 2006.

S. Caille, E. F. Espejo, J. P. Reneric, M. Cador, G. F. Koob et al., Total neurochemical lesion of noradrenergic neurons of the locus ceruleus does not alter either naloxone-precipitated or spontaneous opiate withdrawal nor does it influence ability of clonidine to reverse opiate withdrawal, J Pharmacol Exp Ther, vol.290, pp.881-892, 1999.

S. Candeletti, G. Lopetuso, R. Cannarsa, C. Cavina, and P. Romualdi, Effects of prolonged treatment with the opiate tramadol on prodynorphin gene expression in rat CNS, J Mol Neurosci, vol.30, pp.341-347, 2006.

N. S. Canteras, R. B. Simerly, and L. W. Swanson, Organization of projections from the ventromedial nucleus of the hypothalamus: a Phaseolus vulgaris-leucoagglutinin study in the rat, J Comp Neurol, vol.348, pp.41-79, 1994.

R. N. Cardinal, J. A. Parkinson, J. Hall, and B. J. Everitt, Emotion and motivation: the role of the amygdala, ventral striatum, and prefrontal cortex, Neurosci Biobehav Rev, vol.26, pp.321-352, 2002.

W. A. Carlezon and M. J. Thomas, Biological substrates of reward and aversion: a nucleus accumbens activity hypothesis, Neuropharmacology, vol.56, issue.1, pp.122-132, 2009.

K. D. Carr, D. O. Aleman, T. H. Bak, and E. J. Simon, Effects of parabrachial opioid antagonism on stimulation-induced feeding, Brain Res, vol.545, pp.283-286, 1991.

M. D. Cassell, L. J. Freedman, and C. Shi, The intrinsic organization of the central extended amygdala, Ann NY Acad Sci, vol.877, pp.217-241, 1999.

A. Castane, P. Robledo, A. Matifas, B. L. Kieffer, and R. Maldonado, Cannabinoid withdrawal syndrome is reduced in double mu and delta opioid receptor knockout mice, Eur J Neurosci, vol.17, pp.155-159, 2003.

M. P. Castelli, M. Melis, M. Mameli, P. Fadda, G. Diaz et al., Chronic morphine and naltrexone fail to modify mu-opioid receptor mRNA levels in the rat brain, Brain Res, vol.45, pp.149-153, 1997.

P. Cazala, Dose-dependent effects of morphine differentiate self-administration elicited from lateral hypothalamus and mesencephalic central gray area in mice, Brain Res, vol.527, pp.280-285, 1990.

P. Cazala, C. Darracq, and M. Saint-marc, Self-administration of morphine into the lateral hypothalamus in the mouse, Brain Res, vol.416, pp.283-288, 1987.

P. Cazala, A. Norena, L. Merrer, J. Galey, and D. , Differential involvement of the lateral and medial divisions of the septal area on spatial learning processes as revealed by intracranial selfadministration of morphine in mice, Behav Brain Res, vol.97, pp.179-188, 1998.

G. Q. Chang, O. Karatayev, R. Ahsan, N. M. Avena, C. Lee et al., Effect of ethanol on hypothalamic opioid peptides, enkephalin, and dynorphin: relationship with circulating triglycerides, Alcohol Clin Exp Res, vol.31, pp.249-259, 2007.

V. I. Chefer and T. S. Shippenberg, Augmentation of morphine-induced sensitization but reduction in morphine tolerance and reward in delta-opioid receptor knockout mice, Neuropsychopharmacology, vol.34, pp.887-898, 2009.

M. J. Christie, J. T. Williams, P. B. Osborne, and C. E. Bellchambers, Where is the locus in opioid withdrawal?, Trends Pharmacol Sci, vol.18, pp.134-140, 1997.

S. Collins, D. 'addario, C. Romualdi, P. Candeletti, S. Izenwasser et al., Regulation of dynorphin gene expression by kappa-opioid agonist treatment, Neuroreport, vol.13, pp.107-109, 2002.

A. Contarino, R. Picetti, H. W. Matthes, G. F. Koob, B. L. Kieffer et al., Lack of reward and locomotor stimulation induced by heroin in mu-opioid receptor-deficient mice, Eur J Pharmacol, vol.446, pp.103-109, 2002.

C. Contet, B. L. Kieffer, and K. Befort, Mu opioid receptor: a gateway to drug addiction, Curr Opin Neurobiol, vol.14, pp.370-378, 2004.

L. M. Coolen, J. Allard, W. A. Truitt, and K. E. Mckenna, Central regulation of ejaculation, Physiol Behav, vol.83, pp.203-215, 2004.

J. Corchero, M. A. Avila, J. A. Fuentes, and J. Manzanares, delta-9-Tetrahydrocannabinol increases prodynorphin and proenkephalin gene expression in the spinal cord of the rat, Life Sci, vol.61, pp.39-43, 1997.

J. Corchero, J. A. Fuentes, and J. Manzanares, delta 9-Tetrahydrocannabinol increases proopiomelanocortin gene expression in the arcuate nucleus of the rat hypothalamus, Eur J Pharmacol, vol.323, pp.193-195, 1997.

J. Corchero, J. A. Fuentes, and J. Manzanares, Gender differences in proenkephalin gene expression response to delta9-tetrahydrocan-nabinol in the hypothalamus of the rat, J Psychopharmacol, vol.16, pp.283-289, 2002.

J. Corchero, J. Manzanares, and J. A. Fuentes, Repeated administration of delta9-tetrahydrocannabinol produces a differential time related responsiveness on proenkephalin, proopiomelanocortin and corticotropin releasing factor gene expression in the hypothalamus and pituitary gland of the rat, Neuropharmacology, vol.38, pp.433-439, 1999.

J. Corchero, J. Romero, F. Berrendero, J. Fernandez-ruiz, J. A. Ramos et al., Time-dependent differences of repeated administration with delta9-tetrahydrocannabinol in proenkephalin and cannabinoid receptor gene expression and G-protein activation by mu-opioid and CB1-cannabinoid receptors in the caudate-putamen, Brain Res, vol.67, pp.148-157, 1999.

J. L. Cornish, J. M. Lontos, K. J. Clemens, and I. S. Mcgregor, Cocaine and heroin ("speedball") selfadministration: the involvement of nucleus accumbens dopamine and mu-opiate, but not deltaopiate receptors, Psychopharmacology, vol.180, pp.21-32, 2005.

W. A. Corrigall, Heroin self-administration: effects of antagonist treatment in lateral hypothalamus, Pharmacol Biochem Behav, vol.27, pp.693-700, 1987.

W. A. Corrigall, K. M. Coen, K. L. Adamson, and B. L. Chow, Manipulations of mu-opioid and nicotinic cholinergic receptors in the pontine tegmental region alter cocaine self-administration in rats, Psychopharmacology, vol.145, pp.412-417, 1999.

W. A. Corrigall, K. M. Coen, K. L. Adamson, B. L. Chow, and J. Zhang, Response of nicotine selfadministration in the rat to manipulations of mu-opioid and gamma-aminobutyric acid receptors in the ventral tegmental area, Psychopharmacology, vol.149, pp.107-114, 2000.

W. A. Corrigall, K. M. Coen, J. Zhang, and L. Adamson, Pharmacological manipulations of the pedunculopontine tegmental nucleus in the rat reduce self-administration of both nicotine and cocaine, Psychopharmacology, vol.160, pp.198-205, 2002.

M. S. Cowen and A. J. Lawrence, Alterations in central preproenkephalin mRNA expression after chronic free-choice ethanol consumption by fawn-hooded rats, Alcohol Clin Exp Res, vol.25, pp.1126-1133, 2001.

J. C. Crabbe, Review: neurogenetic studies of alcohol addiction, Philos Trans R Soc Lond B Biol Sci, vol.363, pp.3201-3211, 2008.

J. A. Crespo, J. Manzanares, J. M. Oliva, J. Corchero, T. Palomo et al., Extinction of cocaine self-administration produces a differential time-related regulation of proenkephalin gene expression in rat brain, Neuropsychopharmacology, vol.25, pp.185-194, 2001.

A. Dahlstrom and K. Fuxe, Localization of monoamines in the lower brain stem, Experientia, vol.20, pp.398-399, 1964.

J. E. Dallimore, A. L. Mickiewicz, and T. C. Napier, Intra-ventral pallidal glutamate antagonists block expression of morphine-induced place preference, Behav Neurosci, vol.120, pp.1103-1114, 2006.

S. Daumas, A. Betourne, H. Halley, D. P. Wolfer, H. P. Lipp et al., Transient activation of the CA3 Kappa opioid system in the dorsal hippocampus modulates complex memory processing in mice, Neurobiol Learn Mem, vol.88, pp.94-103, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00167915

J. B. Daunais and J. F. Mcginty, Acute and chronic cocaine administration differentially alters striatal opioid and nuclear transcription factor mRNAs, Synapse, vol.18, pp.35-45, 1994.

J. B. Daunais and J. F. Mcginty, Cocaine binges differentially alter striatal preprodynorphin and zif/268 mRNAs, Brain Res, vol.29, pp.201-210, 1995.

J. B. Daunais and J. F. Mcginty, The effects of D1 or D2 dopamine receptor blockade on zif/268 and preprodynorphin gene expression in rat forebrain following a short-term cocaine binge, Brain Res, vol.35, pp.237-248, 1996.

V. David and P. Cazala, Anatomical and pharmacological specificity of the rewarding effect elicited by microinjections of morphine into the nucleus accumbens of mice, Psychopharmacology, vol.150, pp.24-34, 2000.

V. David and P. Cazala, A comparative study of self-administration of morphine into the amygdala and the ventral tegmental area in mice, Behav Brain Res, vol.65, pp.205-211, 1994.

V. David, T. P. Durkin, and P. Cazala, Differential effects of the dopamine D2/D3 receptor antagonist sulpiride on self-administration of morphine into the ventral tegmental area or the nucleus accumbens, Psychopharmacology, vol.160, pp.307-317, 2002.

V. David, A. Matifas, S. Gavello-baudy, L. Decorte, B. L. Kieffer et al., Brain regional Fos expression elicited by the activation of mu-but not delta-opioid receptors of the ventral tegmental area: evidence for an implication of the ventral thalamus in opiate reward, Neuropsychopharmacology, vol.33, pp.1746-1759, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00282970

J. S. De-olmos and L. Heimer, The concepts of the ventral striatopallidal system and extended amygdala, Ann NY Acad Sci, vol.877, pp.1-32, 1999.

J. M. Delfs, Y. Zhu, J. P. Druhan, A. , and G. , Noradrenaline in the ventral forebrain is critical for opiate withdrawal-induced aversion, Nature, vol.403, pp.430-434, 2000.

A. M. Depaoli, K. M. Hurley, K. Yasada, T. Reisine, and G. Bell, Distribution of kappa opioid receptor mRNA in adult mouse brain: an in situ hybridization histochemistry study, Mol Cell Neurosci, vol.5, pp.327-335, 1994.

V. Deroche-gamonet, D. Belin, and P. V. Piazza, Evidence for addiction-like behavior in the rat, Science, vol.305, pp.1014-1017, 2004.

D. Chiara, G. Imperato, and A. , Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats, Proc Natl Acad Sci USA, vol.85, pp.5274-5278, 1988.

A. H. Dickenson and B. L. Kieffer, Opiates: basic mechanisms, Textbook of Pain, pp.427-442, 2005.

R. J. Dileone, D. Georgescu, and E. J. Nestler, Lateral hypothalamic neuropeptides in reward and drug addiction, Life Sci, vol.73, pp.759-768, 2003.

J. M. Dominguez and E. M. Hull, Dopamine, the medial preoptic area, and male sexual behavior, Physiol Behav, vol.86, pp.356-368, 2005.

Z. Dong, H. Han, M. Wang, L. Xu, W. Hao et al., Morphine conditioned place preference depends on glucocorticoid receptors in both hippocampus and nucleus accumbens, Hippocampus, vol.16, pp.809-813, 2006.

C. T. Drake, C. Chavkin, and T. A. Milner, Opioid systems in the dentate gyrus, Prog Brain Res, vol.163, pp.245-263, 2007.

G. Drolet, E. J. Van-bockstaele, A. , and G. , Robust enkephalin innervation of the locus coeruleus from the rostral medulla, J Neurosci, vol.12, pp.3162-3174, 1992.

F. Ducci and D. Goldman, Genetic approaches to addiction: genes and alcohol, Addiction, vol.103, pp.1414-1428, 2008.

D. Economidou, Y. Pelloux, T. W. Robbins, J. W. Dalley, and B. J. Everitt, High impulsivity predicts relapse to cocaine-seeking after punishment-induced abstinence, Biol Psychiatry, vol.65, pp.851-856, 2009.

M. Ellgren, S. M. Spano, and Y. L. Hurd, Adolescent cannabis exposure alters opiate intake and opioid limbic neuronal populations in adult rats, Neuropsychopharmacology, vol.32, pp.607-615, 2007.

D. H. Epstein, K. L. Preston, J. Stewart, and Y. Shaham, Toward a model of drug relapse: an assessment of the validity of the reinstatement procedure, Psychopharmacology, vol.189, pp.1-16, 2006.

B. J. Everitt and T. W. Robbins, Neural systems of reinforcement for drug addiction: from actions to habits to compulsion, Nat Neurosci, vol.8, pp.1481-1489, 2005.

B. J. Everitt and T. W. Robbins, Second-order schedules of drug reinforcement in rats and monkeys: measurement of reinforcing efficacy and drug-seeking behaviour, Psychopharmacology, vol.153, pp.17-30, 2000.

J. H. Fallon and F. M. Leslie, Distribution of dynorphin and enkephalin peptides in the rat brain, J Comp Neurol, vol.249, pp.293-336, 1986.

Y. Fang, M. J. Kelly, and O. K. Ronnekleiv, Proopiomelanocortin (POMC) mRNA expression: distribution and region-specific downregulation by chronic morphine in female guinea pig hypothalamus, Brain Res, vol.55, pp.1-8, 1998.

D. Filliol, S. Ghozland, J. Chluba, M. Martin, H. W. Matthes et al., Mice deficient for delta-and mu-opioid receptors exhibit opposing alterations of emotional responses, Nat Genet, vol.25, pp.195-200, 2000.

L. M. Flanagan-cato, Estrogen-induced remodeling of hypothalamic neural circuitry, Front Neuroendocrinol, vol.21, pp.309-329, 2000.

K. L. Foster, P. F. Mckay, R. Seyoum, D. Milbourne, W. Yin et al., GABA(A) and opioid receptors of the central nucleus of the amygdala selectively regulate ethanol-maintained behaviors, Neuropsychopharmacology, vol.29, pp.269-284, 2004.

J. S. Fowler, N. D. Volkow, C. A. Kassed, and L. Chang, Imaging the addicted human brain, Sci Pract Perspect, vol.3, pp.4-16, 2007.

Y. Fukunaga, N. Inoue, M. Miyamoto, S. Kishioka, and H. Yamamoto, Effects of peptidase inhibitors, [d-Ala2,Met5]-enkephalinamide and antiserum to methionine-enkephalin microinjected into the caudal periaqueductal gray on morphine withdrawal in rats, Jpn J Pharmacol, vol.78, pp.455-461, 1998.

Y. Fukunaga, S. Nishida, N. Inoue, S. Kishioka, and H. Yamamoto, Increase of preproenkephalin mRNA in the caudal part of peri-aqueductal gray by morphine withdrawal in rats: a quantitative in situ hybridization study, Brain Res, vol.42, pp.128-130, 1996.

C. A. Gadd, P. Murtra, C. De-felipe, and S. P. Hunt, Neurokinin-1 receptor-expressing neurons in the amygdala modulate morphine reward and anxiety behaviors in the mouse, J Neurosci, vol.23, pp.8271-8280, 2003.

L. Galeote, F. Berrendero, A. Bura, S. Zimmer, A. Maldonado et al., Prodynorphin gene disruption increases the sensitivity to nicotine self-administration in mice, Int J Neuropsychopharmacol, pp.1-11, 2008.

M. M. Garcia, H. E. Brown, and R. E. Harlan, Alterations in immediate-early gene proteins in the rat forebrain induced by acute morphine injection, Brain Res, vol.692, pp.23-40, 1995.

E. Garcia-de-yebenes and G. Pelletier, Opioid regulation of proopiomelanocortin (POMC) gene expression in the rat brain as studied by in situ hybridization, Neuropeptides, vol.25, pp.91-94, 1993.

S. P. Garcia-horsman, A. Agmo, and R. G. Paredes, Infusions of naloxone into the medial preoptic area, ventromedial nucleus of the hypothalamus, and amygdala block conditioned place preference induced by paced mating behavior, Horm Behav, vol.54, pp.709-716, 2008.

C. Gaveriaux-ruff and B. L. Kieffer, Conditional gene targeting in the mouse nervous system: insights into brain function and diseases, Pharmacol Ther, vol.113, pp.619-634, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00190962

C. Gaveriaux-ruff and B. L. Kieffer, Opioid receptor genes inactivated in mice: the highlights, Neuropeptides, vol.36, pp.62-71, 2002.

S. R. George, R. L. Zastawny, R. Briones-urbina, R. Cheng, T. Nguyen et al., Distinct distributions of mu, delta and kappa opioid receptor mRNA in rat brain, Biochem Biophys Res Commun, vol.205, pp.1438-1444, 1994.

F. Georges, L. Stinus, B. Bloch, L. Moine, and C. , Chronic morphine exposure and spontaneous withdrawal are associated with modifications of dopamine receptor and neuropeptide gene expression in the rat striatum, Eur J Neurosci, vol.11, pp.481-490, 1999.

M. A. Gerrits, H. B. Lesscher, and J. M. Van-ree, Drug dependence and the endogenous opioid system, Eur Neuropsychopharmacol, vol.13, pp.424-434, 2003.

S. Ghozland, K. Chu, B. L. Kieffer, and A. J. Roberts, Lack of stimulant and anxiolytic-like effects of ethanol and accelerated development of ethanol dependence in mu-opioid receptor knockout mice, Neuropharmacology, vol.49, pp.493-501, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00187545

S. Ghozland, H. W. Matthes, F. Simonin, D. Filliol, B. L. Kieffer et al., Motivational effects of cannabinoids are mediated by mu-opioid and kappa-opioid receptors, J Neurosci, vol.22, pp.1146-1154, 2002.

S. Q. Giraudo, C. J. Billington, and A. S. Levine, Effects of the opioid antagonist naltrexone on feeding induced by DAMGO in the central nucleus of the amygdala and in the paraventricular nucleus in the rat, Brain Res, vol.782, pp.18-23, 1998.

F. Giuliano and O. Rampin, Central neural regulation of penile erection, Neurosci Biobehav Rev, vol.24, pp.517-533, 2000.
URL : https://hal.archives-ouvertes.fr/hal-02689207

B. Givens and M. Sarter, Modulation of cognitive processes by transsynaptic activation of the basal forebrain, Behav Brain Res, vol.84, pp.1-22, 1997.

M. J. Glass, C. J. Billington, and A. S. Levine, Naltrexone administered to central nucleus of amygdala or PVN: neural dissociation of diet and energy, Am J Physiol Regul Integr Comp Physiol, vol.279, pp.86-92, 2000.

M. J. Glass, C. J. Billington, and A. S. Levine, Opioids and food intake: distributed functional neural pathways?, Neuropeptides, vol.33, pp.360-368, 1999.

M. J. Glass, J. E. Briggs, C. J. Billington, C. M. Kotz, and A. S. Levine, Opioid receptor blockade in rat nucleus tractus solitarius alters amygdala dynorphin gene expression, Am J Physiol Regul Integr Comp Physiol, vol.283, pp.161-167, 2002.

N. E. Goeders, J. D. Lane, and J. E. Smith, Self-administration of methionine enkephalin into the nucleus accumbens, Pharmacol Biochem Behav, vol.20, pp.451-455, 1984.

J. Gong, X. W. Li, Z. Lai, J. C. Froehlich, and L. Yu, Quantitative comparison of mu opioid receptor mRNA in selected CNS regions of alcohol naive rats selectively bred for high and low alcohol drinking, Neurosci Lett, vol.227, pp.9-12, 1997.

S. Gong, C. Zheng, M. L. Doughty, K. Losos, N. Didkovsky et al., A gene expression atlas of the central nervous system based on bacterial artificial chromosomes, Nature, vol.425, pp.917-925, 2003.

W. Gong, D. Neill, and J. B. Justice, Conditioned place preference and locomotor activation produced by injection of psychostimulants into ventral pallidum, Brain Res, vol.707, pp.64-74, 1996.

R. J. Goody, S. M. Oakley, D. Filliol, B. L. Kieffer, and I. Kitchen, Quantitative autoradiographic mapping of opioid receptors in the brain of delta-opioid receptor gene knockout mice, Brain Res, vol.945, pp.9-19, 2002.

K. P. Gudehithlu and H. N. Bhargava, Modulation of preproenkephalin mRNA levels in brain regions and spinal cord of rats treated chronically with morphine, Peptides, vol.16, pp.415-419, 1995.

K. Gulya, A. K. Orpana, J. M. Sikela, and P. L. Hoffman, Prodynorphin and vasopressin mRNA levels are differentially affected by chronic ethanol ingestion in the mouse, Brain Res, vol.20, pp.1-8, 1993.

N. Guo, M. M. Garcia, B. K. Taylor, J. E. Zadina, and R. E. Harlan, Blockade of micro-opioid receptors in the medial thalamus inhibits acquisition, but not expression, of morphine-induced conditioned place preference, Neuroscience, vol.151, pp.948-954, 2008.

F. S. Hall, M. Goeb, X. F. Li, I. Sora, and G. R. Uhl, mu-Opioid receptor knockout mice display reduced cocaine conditioned place preference but enhanced sensitization of cocaine-induced locomotion, Brain Res, vol.121, pp.123-130, 2004.

M. Harbuz, J. A. Russell, B. E. Sumner, M. Kawata, and S. L. Lightman, Rapid changes in the content of proenkephalin A and corticotrophin releasing hormone mRNAs in the paraventricular nucleus during morphine withdrawal in urethane-anaesthetized rats, Brain Res, vol.9, pp.285-291, 1991.

R. E. Harlan, B. D. Shivers, G. J. Romano, R. D. Howells, and D. W. Pfaff, Localization of preproenkephalin mRNA in the rat brain and spinal cord by in situ hybridization, J Comp Neurol, vol.258, pp.159-184, 1987.

G. C. Harris, A. , and G. , Arousal and reward: a dichotomy in orexin function, Trends Neurosci, vol.29, pp.571-577, 2006.

M. D. Hayward, S. T. Hansen, J. E. Pintar, and M. J. Low, Operant self-administration of ethanol in C57BL/6 mice lacking beta-endorphin and enkephalin, Pharmacol Biochem Behav, vol.79, pp.171-181, 2004.

M. D. Hayward, A. Schaich-borg, J. E. Pintar, and M. J. Low, Differential involvement of endogenous opioids in sucrose consumption and food reinforcement, Pharmacol Biochem Behav, vol.85, pp.601-611, 2006.

L. A. Herraez-baranda, J. Carretero, R. Gonzalez-sarmiento, and R. E. Rodriguez, Kappa opioid receptor is expressed in the rat cerebellar cortex, Cell Tissue Res, vol.320, pp.223-228, 2005.

C. J. Heyser, A. J. Roberts, G. Schulteis, and G. F. Koob, Central administration of an opiate antagonist decreases oral ethanol self-administration in rats, Alcohol Clin Exp Res, vol.23, pp.1468-1476, 1999.

S. Higuchi, S. Matsushita, and H. Kashima, New findings on the genetic influences on alcohol use and dependence, Curr Opin Psychiatry, vol.19, pp.253-265, 2006.

N. Hiroi and N. M. White, The ventral pallidum area is involved in the acquisition but not expression of the amphetamine conditioned place preference, Neurosci Lett, vol.156, pp.9-12, 1993.

P. C. Holland and M. Gallagher, Amygdala-frontal interactions and reward expectancy, Curr Opin Neurobiol, vol.14, pp.148-155, 2004.

P. C. Holland, G. D. Petrovich, and M. Gallagher, The effects of amygdala lesions on conditioned stimulus-potentiated eating in rats, Physiol Behav, vol.76, pp.117-129, 2002.

V. Hollt and I. Haarmann, Differential alterations by chronic treatment with morphine of proopiomelanocortin mRNA levels in anterior as compared to intermediate pituitary lobes of rats, Neuropeptides, vol.5, pp.481-484, 1985.

V. Hollt and G. Horn, Effect of nicotine on mRNA levels encoding opioid peptides, vasopressin and alpha 3 nicotinic receptor subunit in the rat, Clin Invest, vol.70, pp.224-231, 1992.

A. A. Houdi, R. Dasgupta, and M. S. Kindy, Effect of nicotine use and withdrawal on brain preproenkephalin A mRNA, Brain Res, vol.799, pp.257-263, 1998.

H. Houshyar, S. Manalo, and M. F. Dallman, Time-dependent alterations in mRNA expression of brain neuropeptides regulating energy balance and hypothalamo-pituitary-adrenal activity after withdrawal from intermittent morphine treatment, J Neurosci, vol.24, pp.9414-9424, 2004.

L. Z. Huang and U. H. Winzer-serhan, Nicotine regulates mRNA expression of feeding peptides in the arcuate nucleus in neonatal rat pups, Dev Neurobiol, vol.67, pp.363-377, 2007.

C. B. Hubner and G. F. Koob, The ventral pallidum plays a role in mediating cocaine and heroin selfadministration in the rat, Brain Res, vol.508, pp.20-29, 1990.

E. M. Hull and J. M. Dominguez, Sexual behavior in male rodents, Horm Behav, vol.52, pp.45-55, 2007.

Y. L. Hurd, E. E. Brown, J. M. Finlay, H. C. Fibiger, and C. R. Gerfen, Cocaine self-administration differentially alters mRNA expression of striatal peptides, Brain Res, vol.13, pp.165-170, 1992.

S. E. Hyman, R. C. Malenka, and E. J. Nestler, Neural mechanisms of addiction: the role of reward-related learning and memory, Annu Rev Neurosci, vol.29, pp.565-598, 2006.

P. Hyytia and K. Kiianmaa, Suppression of ethanol responding by centrally administered CTOP and naltrindole in AA and Wistar rats, Alcohol Clin Exp Res, vol.25, pp.25-33, 2001.

K. Ikeda, S. Ide, W. Han, M. Hayashida, G. R. Uhl et al., How individual sensitivity to opiates can be predicted by gene analyses, Trends Pharmacol Sci, vol.26, pp.311-317, 2005.

S. Ikemoto and R. A. Wise, Mapping of chemical trigger zones for reward, Neuropharmacology, vol.47, issue.1, pp.190-201, 2004.

R. Isola, H. Zhang, A. M. Duchemin, G. A. Tejwani, N. H. Neff et al., Met-enkephalin and preproenkephalin mRNA changes in the striatum of the nicotine abstinence mouse, Neurosci Lett, vol.325, pp.67-71, 2002.

R. Isola, H. Zhang, G. A. Tejwani, N. H. Neff, and M. Hadjiconstantinou, Dynorphin and prodynorphin mRNA changes in the striatum during nicotine withdrawal, Synapse, vol.62, pp.448-455, 2008.

R. Ito, T. W. Robbins, C. M. Pennartz, and B. J. Everitt, Functional interaction between the hippocampus and nucleus accumbens shell is necessary for the acquisition of appetitive spatial context conditioning, J Neurosci, vol.28, pp.6950-6959, 2008.

F. Jenck, A. Gratton, and R. A. Wise, Opioid receptor subtypes associated with ventral tegmental facilitation of lateral hypothalamic brain stimulation reward, Brain Res, vol.423, pp.34-38, 1987.

C. Jomary, J. E. Gairin, J. Cros, and J. C. Meunier, Autoradiographic localization of supraspinal kappaopioid receptors with [ 125 I-Tyr1, d-Pro10]dynorphin A-(1-11), Proc Natl Acad Sci USA, vol.85, pp.627-631, 1988.

K. L. Jones and G. A. Barr, Injections of an opioid antagonist into the locus coeruleus and periaqueductal gray but not the amygdala precipitates morphine withdrawal in the 7-day-old rat, Synapse, vol.39, pp.139-151, 2001.

H. L. June, R. Cummings, . Eiler, K. L. Foster, P. F. Mckay et al., Central opioid receptors differentially regulate the nalmefeneinduced suppression of ethanol-and saccharin-reinforced behaviors in alcohol-preferring (P) rats, Neuropsychopharmacology, vol.29, pp.285-299, 2004.

P. W. Kalivas and K. Mcfarland, Brain circuitry and the reinstatement of cocaine-seeking behavior, Psychopharmacology, vol.168, pp.44-56, 2003.

M. J. Kas, R. Van-den-bos, A. M. Baars, M. Lubbers, H. M. Lesscher et al., Mu-opioid receptor knockout mice show diminished food-anticipatory activity, Eur J Neurosci, vol.20, pp.1624-1632, 2004.

J. L. Katz and S. T. Higgins, The validity of the reinstatement model of craving and relapse to drug use, Psychopharmacology, vol.168, pp.21-30, 2003.

T. Kawagoe, R. Tamura, T. Uwano, T. Asahi, H. Nishijo et al., Neural correlates of stimulus-reward association in the rat mediodorsal thalamus, Neuroreport, vol.18, pp.683-688, 2007.

A. E. Kelley, Memory and addiction: shared neural circuitry and molecular mechanisms, Neuron, vol.44, pp.161-179, 2004.

A. E. Kelley, V. P. Bakshi, S. N. Haber, T. L. Steininger, M. J. Will et al., Opioid modulation of taste hedonics within the ventral striatum, Physiol Behav, vol.76, pp.365-377, 2002.

A. E. Kelley, B. A. Baldo, and W. E. Pratt, A proposed hypothalamic-thalamic-striatal axis for the integration of energy balance, arousal, and food reward, J Comp Neurol, vol.493, pp.72-85, 2005.

A. E. Kelley and K. C. Berridge, The neuroscience of natural rewards: relevance to addictive drugs, J Neurosci, vol.22, pp.3306-3311, 2002.

H. Khachaturian, M. E. Lewis, S. N. Haber, R. A. Houghten, H. Akil et al., Prodynorphin peptide immunocytochemistry in rhesus monkey brain, Peptides, vol.6, issue.2, pp.155-166, 1985.

H. Khachaturian, M. E. Lewis, and S. J. Watson, Enkephalin systems in diencephalon and brainstem of the rat, J Comp Neurol, vol.220, pp.310-320, 1983.

H. Khachaturian, S. J. Watson, M. E. Lewis, D. Coy, A. Goldstein et al., Dynorphin immunocytochemistry in the rat central nervous system, Peptides, vol.3, pp.941-954, 1982.

B. L. Kieffer, Recent advances in molecular recognition and signal transduction of active peptides: receptors for opioid peptides, Cell Mol Neurobiol, vol.15, pp.615-635, 1995.

B. L. Kieffer and C. Gaveriaux-ruff, Exploring the opioid system by gene knockout, Prog Neurobiol, vol.66, pp.285-306, 2002.

E. M. Kim, J. G. Quinn, A. S. Levine, and E. O'hare, A bi-directional mu-opioid-opioid connection between the nucleus of the accumbens shell and the central nucleus of the amygdala in the rat, Brain Res, vol.1029, pp.135-139, 2004.

I. Kitchen, S. J. Slowe, H. W. Matthes, and B. Kieffer, Quantitative autoradiographic mapping of mu-, delta-and kappa-opioid receptors in knockout mice lacking the mu-opioid receptor gene, Brain Res, vol.778, pp.73-88, 1997.

H. N. Koenig and M. F. Olive, Ethanol consumption patterns and conditioned place preference in mice lacking preproenkephalin, Neurosci Lett, vol.325, pp.75-78, 2002.

M. D. Kohnke, Approach to the genetics of alcoholism: a review based on pathophysiology, Biochem Pharmacol, vol.75, pp.160-177, 2008.

G. Koob and M. J. Kreek, Stress, dysregulation of drug reward pathways, and the transition to drug dependence, Am J Psychiatry, vol.164, pp.1149-1159, 2007.

G. F. Koob, Drugs of abuse: anatomy, pharmacology and function of reward pathways, Trends Pharmacol Sci, vol.13, pp.177-184, 1992.

G. F. Koob, Neuroadaptive mechanisms of addiction: studies on the extended amygdala, Eur Neuropsychopharmacol, vol.13, pp.442-452, 2003.

G. F. Koob, Neurobiological substrates for the dark side of compulsivity in addiction, Neuropharmacology, vol.56, issue.1, pp.18-31, 2009.

G. F. Koob, A role for brain stress systems in addiction, Neuron, vol.59, pp.11-34, 2008.

G. F. Koob, L. Moal, and M. , Addiction and the brain antireward system, Annu Rev Psychol, vol.59, pp.29-53, 2008.

G. F. Koob, L. Moal, and M. , Drug addiction, dysregulation of reward, and allostasis, Neuropsychopharmacology, vol.24, pp.97-129, 2001.

G. F. Koob, T. L. Wall, and F. E. Bloom, Nucleus accumbens as a substrate for the aversive stimulus effects of opiate withdrawal, Psychopharmacology, vol.98, pp.530-534, 1989.

K. M. Kovacs, I. Szakall, D. O'brien, R. Wang, K. Y. Vinod et al., Decreased oral self-administration of alcohol in kappa-opioid receptor knock-out mice, Alcohol Clin Exp Res, vol.29, pp.730-738, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00129902

M. J. Kreek, G. Bart, C. Lilly, K. S. Laforge, and D. A. Nielsen, Pharmacogenetics and human molecular genetics of opiate and cocaine addictions and their treatments, Pharmacol Rev, vol.57, pp.1-26, 2005.

M. J. Kreek, Y. Zhou, E. R. Butelman, and O. Levran, Opiate and cocaine addiction: from bench to clinic and back to the bench, Curr Opin Pharmacol, vol.9, pp.74-80, 2009.

B. B. Land, M. R. Bruchas, J. C. Lemos, M. Xu, E. J. Melief et al., The dysphoric component of stress is encoded by activation of the dynorphin kappa-opioid system, J Neurosci, vol.28, pp.407-414, 2008.

T. A. Lantos, T. J. Gorcs, and M. Palkovits, Immunohistochemical mapping of neuropeptides in the premamillary region of the hypothalamus in rats, Brain Res, vol.20, pp.209-249, 1995.

L. Merrer, J. Gavello-baudy, S. Galey, D. Cazala, and P. , Morphine self-administration into the lateral septum depends on dopaminergic mechanisms: evidence from pharmacology and Fos neuroimaging, Behav Brain Res, vol.180, pp.203-217, 2007.

F. Leri, Y. Zhou, B. Goddard, E. Cummins, and M. J. Kreek, Effects of high-dose methadone maintenance on cocaine place conditioning, cocaine self-administration, and mu-opioid receptor mRNA expression in the rat brain, Neuropsychopharmacology, vol.31, pp.1462-1474, 2006.

M. Leriche, A. Cote-velez, and M. Mendez, Presence of pro-opiomelanocortin mRNA in the rat medial prefrontal cortex, nucleus accumbens and ventral tegmental area: studies by RT-PCR and in situ hybridization techniques, Neuropeptides, vol.41, pp.421-431, 2007.

H. M. Lesscher, A. Bailey, J. P. Burbach, J. M. Van-ree, I. Kitchen et al., Receptor-selective changes in mu-, delta-and kappa-opioid receptors after chronic naltrexone treatment in mice, Eur J Neurosci, vol.17, pp.1006-1012, 2003.

A. S. Levine, P. K. Olszewski, M. A. Mullett, J. D. Pomonis, M. K. Grace et al., Intraamygdalar injection of DAMGO: effects on c-Fos levels in brain sites associated with feeding behavior, Brain Res, vol.1015, pp.9-14, 2004.

M. Leyton and J. Stewart, The stimulation of central kappa opioid receptors decreases male sexual behavior and locomotor activity, Brain Res, vol.594, pp.56-74, 1992.

D. Li, P. K. Olszewski, Q. Shi, M. K. Grace, C. J. Billington et al., Effect of opioid receptor ligands injected into the rostral lateral hypothalamus on c-fos and feeding behavior, Brain Res, vol.1096, pp.120-124, 2006.

M. D. Li, O. Konu, J. K. Kane, and K. G. Becker, Microarray technology and its application on nicotine research, Mol Neurobiol, vol.25, pp.265-285, 2002.

J. Liang, Y. Li, X. Ping, P. Yu, Y. Zuo et al., The possible involvement of endogenous ligands for mu-, delta-and kappa-opioid receptors in modulating morphine-induced CPP expression in rats, Peptides, vol.27, pp.3307-3314, 2006.

A. H. Lichtman, S. M. Sheikh, H. H. Loh, and B. R. Martin, Opioid and cannabinoid modulation of precipitated withdrawal in delta(9)-tetrahydrocannabinol and morphine-dependent mice, J Pharmacol Exp Ther, vol.298, pp.1007-1014, 2001.

S. L. Lightman and . Young, Corticotrophin-releasing factor, vasopressin and proopiomelanocortin mRNA responses to stress and opiates in the rat, J Physiol, vol.403, pp.511-523, 1988.

A. F. Macdonald, C. J. Billington, and A. S. Levine, Alterations in food intake by opioid and dopamine signaling pathways between the ventral tegmental area and the shell of the nucleus accumbens, Brain Res, vol.1018, pp.78-85, 2004.

A. F. Macdonald, C. J. Billington, and A. S. Levine, Effects of the opioid antagonist naltrexone on feeding induced by DAMGO in the ventral tegmental area and in the nucleus accumbens shell region in the rat, Am J Physiol Regul Integr Comp Physiol, vol.285, pp.999-1004, 2003.

K. Magendzo and G. Bustos, Expression of amphetamine-induced behavioral sensitization after shortand long-term withdrawal periods: participation of mu-and delta-opioid receptors, Neuropsychopharmacology, vol.28, pp.468-477, 2003.

P. Mailleux and J. J. Vanderhaeghen, ?-9-Tetrahydrocannabinol regulates substance P and enkephalin mRNAs levels in the caudateputamen, Eur J Pharmacol, vol.267, pp.1-3, 1994.

R. Maldonado, Participation of noradrenergic pathways in the expression of opiate withdrawal: biochemical and pharmacological evidence, Neurosci Biobehav Rev, vol.21, pp.91-104, 1997.

R. Maldonado, L. Stinus, L. H. Gold, and G. F. Koob, Role of different brain structures in the expression of the physical morphine withdrawal syndrome, J Pharmacol Exp Ther, vol.261, pp.669-677, 1992.

A. Mansour, C. A. Fox, H. Akil, and S. J. Watson, Opioid-receptor mRNA expression in the rat CNS: anatomical and functional implications, Trends Neurosci, vol.18, pp.22-29, 1995.

A. Mansour, C. A. Fox, S. Burke, F. Meng, R. C. Thompson et al., Mu, delta, and kappa opioid receptor mRNA expression in the rat CNS: an in situ hybridization study, J Comp Neurol, vol.350, pp.412-438, 1994.

A. Mansour, M. T. Hoversten, L. P. Taylor, S. J. Watson, and H. Akil, The cloned mu, delta and kappa receptors and their endogenous ligands: evidence for two opioid peptide recognition cores, Brain Res, vol.700, pp.89-98, 1995.

A. Mansour, H. Khachaturian, M. E. Lewis, H. Akil, and S. J. Watson, Autoradiographic differentiation of mu, delta, and kappa opioid receptors in the rat forebrain and midbrain, J Neurosci, vol.7, pp.2445-2464, 1987.

A. Mansour and S. J. Watson, Anatomical distribution of opioid receptors in mammalian: an overview, pp.79-105, 1993.

J. Manzanares, J. Corchero, J. Romero, J. J. Fernandez-ruiz, J. A. Ramos et al., Chronic administration of cannabinoids regulates proenkephalin mRNA levels in selected regions of the rat brain, Brain Res, vol.55, pp.126-132, 1998.

E. B. Margolis, H. L. Fields, G. O. Hjelmstad, and J. M. Mitchell, Delta-opioid receptor expression in the ventral tegmental area protects against elevated alcohol consumption, J Neurosci, vol.28, pp.12672-12681, 2008.

P. Marquez, R. Baliram, N. Gajawada, T. C. Friedman, and K. Lutfy, Differential involvement of enkephalins in analgesic tolerance, locomotor sensitization, and conditioned place preference induced by morphine, Behav Neurosci, vol.120, pp.10-15, 2006.

P. Marquez, R. Baliram, B. L. Kieffer, and K. Lutfy, The mu opioid receptor is involved in buprenorphine-induced locomotor stimulation and conditioned place preference, Neuropharmacology, vol.52, pp.1336-1341, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00189977

T. J. Martin, M. Coller, C. Co, and J. E. Smith, Micro-opioid receptor alkylation in the ventral pallidum and ventral tegmental area, but not in the nucleus accumbens, attenuates the effects of heroin on cocaine self-administration in rats, Neuropsychopharmacology, vol.33, pp.1171-1178, 2008.

T. J. Martin, S. A. Kim, Y. Lyupina, and J. E. Smith, Differential involvement of mu-opioid receptors in the rostral versus caudal nucleus accumbens in the reinforcing effects of heroin in rats: evidence from focal injections of beta-funaltrexamine, Psychopharmacology, vol.161, pp.152-159, 2002.

A. M. Mathieu-kia and M. J. Besson, Repeated administration of cocaine, nicotine and ethanol: effects on preprodynorphin, preprotachykinin A and preproenkephalin mRNA expression in the dorsal and the ventral striatum of the rat, Brain Res, vol.54, pp.141-151, 1998.

A. M. Mathieu, J. Caboche, and M. J. Besson, Distribution of preproenkephalin, preprotachykinin A, and preprodynorphin mRNAs in the rat nucleus accumbens: effect of repeated administration of nicotine, Synapse, vol.23, pp.94-106, 1996.

D. S. Mathon, H. M. Lesscher, M. A. Gerrits, A. Kamal, J. E. Pintar et al., Increased GABAergic input to ventral tegmental area dopaminergic neurons associated with decreased cocaine reinforcement in mu-opioid receptor knockout mice, Neuroscience, vol.130, pp.359-367, 2005.

H. W. Matthes, R. Maldonado, F. Simonin, O. Valverde, S. Slowe et al., Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene, Nature, vol.383, pp.819-823, 1996.

L. Matuszewich, J. L. Ormsby, J. Moses, D. S. Lorrain, and E. M. Hull, Effects of morphiceptin in the medial preoptic area on male sexual behavior, Psychopharmacology, vol.122, pp.330-335, 1995.

P. Mayer and V. Hollt, Pharmacogenetics of opioid receptors and addiction, Pharmacogenet Genomics, vol.16, pp.1-7, 2006.

G. M. Mcalonan, T. W. Robbins, and B. J. Everitt, Effects of medial dorsal thalamic and ventral pallidal lesions on the acquisition of a conditioned place preference: further evidence for the involvement of the ventral striatopallidal system in reward-related processes, Neuroscience, vol.52, pp.605-620, 1993.

W. J. Mcbride, J. M. Murphy, and S. Ikemoto, Localization of brain reinforcement mechanisms: intracranial self-administration and intracranial place-conditioning studies, Behav Brain Res, vol.101, pp.129-152, 1999.

C. A. Mcclung, The molecular mechanisms of morphine addiction, Rev Neurosci, vol.17, pp.393-402, 2006.

A. Mcgregor and J. Herbert, The effects of beta-endorphin infusions into the amygdala on visual and olfactory sensory processing during sexual behaviour in the male rat, Neuroscience, vol.46, pp.173-179, 1992.

A. Mcgregor and J. Herbert, Specific effects of beta-endorphin infused into the amygdala on sexual behaviour in the male rat, Neuroscience, vol.46, pp.165-172, 1992.

K. E. Mckenna, The neurophysiology of female sexual function, World J Urol, vol.20, pp.93-100, 2002.

J. P. Mclaughlin, B. B. Land, S. Li, J. E. Pintar, and C. Chavkin, Prior activation of kappa opioid receptors by U50,488 mimics repeated forced swim stress to potentiate cocaine place preference conditioning, Neuropsychopharmacology, vol.31, pp.787-794, 2006.

J. P. Mclaughlin, M. Marton-popovici, and C. Chavkin, Kappa opioid receptor antagonism and prodynorphin gene disruption block stress-induced behavioral responses, J Neurosci, vol.23, pp.5674-5683, 2003.

I. Merchenthaler, J. L. Maderdrut, P. Cianchetta, P. Shughrue, and D. Bronstein, In situ hybridization histochemical localization of prodynorphin messenger RNA in the central nervous system of the rat, J Comp Neurol, vol.384, pp.211-232, 1997.

C. M. Miranda-paiva, E. R. Ribeiro-barbosa, N. S. Canteras, and L. F. Felicio, A role for the periaqueductal grey in opioidergic inhibition of maternal behaviour, Eur J Neurosci, vol.18, pp.667-674, 2003.

H. Mizoguchi, H. E. Wu, M. Narita, I. Sora, S. F. Hall et al., Lack of mu-opioid receptor-mediated G-protein activation in the spinal cord of mice lacking Exon 1 or Exons 2 and 3 of the MOR-1 gene, J Pharmacol Sci, vol.93, pp.423-429, 2003.

I. Mocchetti and E. Costa, In vivo studies of the regulation of neuropeptide stores in structures of the rat brain, Neuropharmacology, vol.26, pp.855-862, 1987.

I. Mocchetti, A. Ritter, and E. Costa, Down-regulation of proopiomelanocortin synthesis and betaendorphin utilization in hypothalamus of morphine-tolerant rats, J Mol Neurosci, vol.1, pp.33-38, 1989.

A. Moles, B. L. Kieffer, D. Amato, and F. R. , Deficit in attachment behavior in mice lacking the mu-opioid receptor gene, Science, vol.304, pp.1983-1986, 2004.

M. Morales-mulia, F. Panayi, L. Lambas-senas, H. Scarna, and M. Mendez, Changes in proenkephalin mRNA expression in forebrain areas after amphetamine-induced behavioural sensitization, Pharmacol Biochem Behav, vol.87, pp.232-240, 2007.

Y. Morita, J. H. Zhang, T. Hironaka, E. Tateno, K. Noguchi et al., Postnatal development of preproenkephalin mRNA containing neurons in the rat lower brainstem, J Comp Neurol, vol.292, pp.193-213, 1990.

G. J. Morton, R. W. Gelling, K. D. Niswender, C. D. Morrison, C. J. Rhodes et al., Leptin regulates insulin sensitivity via phosphatidylinositol-3-OH kinase signaling in mediobasal hypothalamic neurons, Cell Metab, vol.2, pp.411-420, 2005.

S. Moufid-bellancourt, R. Razafimanalina, and L. Velley, Interaction between mu and kappa receptors located in the parabrachial area in the opioid control of preference threshold for saccharin: modulatory role of lateral hypothalamic neurones, Behav Pharmacol, vol.7, pp.798-809, 1996.
URL : https://hal.archives-ouvertes.fr/hal-02689862

S. Moufid-bellancourt and L. Velley, Effects of morphine injection into the parabrachial area on saccharin preference: modulation by lateral hypothalamic neurons, Pharmacol Biochem Behav, vol.48, pp.127-133, 1994.

E. A. Murray, The amygdala, reward and emotion, Trends Cogn Sci, vol.11, pp.489-497, 2007.

A. M. Naleid, M. K. Grace, M. Chimukangara, C. J. Billington, and A. S. Levine, Paraventricular opioids alter intake of high-fat but not high-sucrose diet depending on diet preference in a binge model of feeding, Am J Physiol Regul Integr Comp Physiol, vol.293, pp.99-105, 2007.

E. E. Nelson and J. Panksepp, Brain substrates of infant-mother attachment: contributions of opioids, oxytocin, and norepinephrine, Neurosci Biobehav Rev, vol.22, pp.437-452, 1998.

E. J. Nestler, Historical review: molecular and cellular mechanisms of opiate and cocaine addiction, Trends Pharmacol Sci, vol.25, pp.210-218, 2004.

E. J. Nestler, Is there a common molecular pathway for addiction?, Nat Neurosci, vol.8, pp.1445-1449, 2005.

D. M. Nicklous and K. J. Simansky, Neuropeptide FF exerts pro-and anti-opioid actions in the parabrachial nucleus to modulate food intake, Am J Physiol Regul Integr Comp Physiol, vol.285, pp.1046-1054, 2003.

K. Niikura, M. Narita, D. Okutsu, Y. Tsurukawa, K. Nanjo et al., Implication of endogenous beta-endorphin in the inhibition of the morphine-induced rewarding effect by the direct activation of spinal protein kinase C in mice, Neurosci Lett, vol.433, pp.54-58, 2008.

R. Norgren, Taste pathways to hypothalamus and amygdala, J Comp Neurol, vol.166, pp.17-30, 1976.

R. D. Oades and G. M. Halliday, Ventral tegmental (A10) system: neurobiology. 1. Anatomy and connectivity, Brain Res, vol.434, pp.117-165, 1987.

J. Olds and P. Milner, Positive reinforcement produced by electrical stimulation of septal area and other regions of rat brain, J Comp Physiol Psychol, vol.47, pp.419-427, 1954.

M. E. Olds, Reinforcing effects of morphine in the nucleus accumbens, Brain Res, vol.237, pp.429-440, 1982.

M. E. Olds and K. N. Williams, Self-administration of d-Ala2-Met-enkephalinamide at hypothalamic self-stimulation sites, Brain Res, vol.194, pp.155-170, 1980.

J. M. Oliva and J. Manzanares, Gene transcription alterations associated with decrease of ethanol intake induced by naltrexone in the brain of Wistar rats, Neuropsychopharmacology, vol.32, pp.1358-1369, 2007.

J. M. Oliva, S. Ortiz, T. Palomo, and J. Manzanares, Behavioural and gene transcription alterations induced by spontaneous cannabinoid withdrawal in mice, J Neurochem, vol.85, pp.94-104, 2003.

C. Olmstead, A. M. Ouaggazzal, and B. L. Kieffer, Deletion of mu opioid receptors in mice decreases impulsivity

M. C. Olmstead and K. B. Franklin, The development of a conditioned place preference to morphine: effects of lesions of various CNS sites, Behav Neurosci, vol.111, pp.1313-1323, 1997.

M. C. Olmstead and K. B. Franklin, The development of a conditioned place preference to morphine: effects of microinjections into various CNS sites, Behav Neurosci, vol.111, pp.1324-1334, 1997.

M. C. Olmstead, E. M. Munn, K. B. Franklin, and R. A. Wise, Effects of pedunculopontine tegmental nucleus lesions on responding for intravenous heroin under different schedules of reinforcement, J Neurosci, vol.18, pp.5035-5044, 1998.

M. C. Olmstead, J. A. Parkinson, F. J. Miles, B. J. Everitt, and A. Dickinson, Cocaine-seeking by rats: regulation, reinforcement and activation, Psychopharmacology, vol.152, pp.123-131, 2000.

V. G. Olson, T. A. Green, R. L. Neve, and E. J. Nestler, Regulation of morphine reward and feeding by CREB in the lateral hypothalamus, Synapse, vol.61, pp.110-113, 2007.

P. K. Olszewski and A. S. Levine, Central opioids and consumption of sweet tastants: when reward outweighs homeostasis, Physiol Behav, vol.91, pp.506-512, 2007.

D. W. Oslin, W. H. Berrettini, O. 'brien, and C. P. , Targeting treatments for alcohol dependence: the pharmacogenetics of naltrexone, Addict Biol, vol.11, pp.397-403, 2006.

J. Panksepp, E. Nelson, and M. Bekkedal, Brain systems for the mediation of social separation-distress and social-reward. Evolutionary antecedents and neuropeptide intermediaries, Ann NY Acad Sci, vol.807, pp.78-100, 1997.

F. Papaleo, B. L. Kieffer, A. Tabarin, and A. Contarino, Decreased motivation to eat in mu-opioid receptor-deficient mice, Eur J Neurosci, vol.25, pp.3398-3405, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00189394

R. G. Paredes, Evaluating the neurobiology of sexual reward, Ilar J, vol.50, pp.15-27, 2008.

S. Pecina and K. C. Berridge, Hedonic hot spot in nucleus accumbens shell: where do mu-opioids cause increased hedonic impact of sweetness?, J Neurosci, vol.25, pp.11777-11786, 2005.

S. Pecina and K. C. Berridge, Opioid site in nucleus accumbens shell mediates eating and hedonic "liking" for food: map based on microinjection Fos plumes, Brain Res, vol.863, pp.71-86, 2000.

S. Pecina, K. S. Smith, and K. C. Berridge, Hedonic hot spots in the brain, Neuroscientist, vol.12, pp.500-511, 2006.

Y. Pelloux, B. J. Everitt, and A. Dickinson, Compulsive drug seeking by rats under punishment: effects of drug taking history, Psychopharmacology, vol.194, pp.127-137, 2007.

H. O. Pettit, A. Ettenberg, F. E. Bloom, and G. F. Koob, Destruction of dopamine in the nucleus accumbens selectively attenuates cocaine but not heroin self-administration in rats, Psychopharmacology, vol.84, pp.167-173, 1984.

J. G. Pfaus and B. J. Everitt, Psychopharmacology: The Forth Generation of Progress, pp.742-758, 1995.

J. G. Pfaus and B. B. Gorzalka, Opioids and sexual behavior, Neurosci Biobehav Rev, vol.11, pp.1-34, 1987.

J. G. Pfaus, T. E. Kippin, and S. Centeno, Conditioning and sexual behavior: a review, Horm Behav, vol.40, pp.291-321, 2001.

B. V. Phillips-farfan, A. Fernandez-guasti, and . Endocrine, neural and pharmacological aspects of sexual satiety in male rats, Neurosci Biobehav Rev, vol.33, pp.442-455, 2009.

J. F. Poulin, Z. Castonguay-lebel, S. Laforest, and G. Drolet, Enkephalin co-expression with classic neurotransmitters in the amygdaloid complex of the rat, J Comp Neurol, vol.506, pp.943-959, 2008.

J. F. Poulin, B. Chevalier, S. Laforest, and G. Drolet, Enkephalinergic afferents of the centromedial amygdala in the rat, J Comp Neurol, vol.496, pp.859-876, 2006.

A. A. Pradhan and P. B. Clarke, Comparison between delta-opioid receptor functional response and autoradiographic labeling in rat brain and spinal cord, J Comp Neurol, vol.481, pp.416-426, 2005.

B. Przewlocka and W. Lason, Adaptive changes in the proenkephalin and D2 dopamine receptor mRNA expression after chronic cocaine in the nucleus accumbens and striatum of the rat, Eur Neuropsychopharmacol, vol.5, pp.465-469, 1995.

B. Przewlocka, W. Lason, and R. Przewlocki, Repeated ethanol administration decreases prodynorphin biosynthesis in the rat hippocampus, Neurosci Lett, vol.134, pp.195-198, 1992.

B. Przewlocka, W. Lason, and R. Przewlocki, Repeated ethanol differently affects opioid peptide biosynthesis in the rat pituitary, Neuroendocrinology, vol.60, pp.331-336, 1994.

B. Przewlocka, J. Turchan, W. Lason, and R. Przewlocki, The effect of single and repeated morphine administration on the prodynorphin system activity in the nucleus accumbens and striatum of the rat, Neuroscience, vol.70, pp.749-754, 1996.

B. Przewlocka, J. Turchan, W. Lason, and R. Przewlocki, Ethanol withdrawal enhances the prodynorphin system activity in the rat nucleus accumbens, Neurosci Lett, vol.238, pp.13-16, 1997.

R. Przewlocki, Opioid abuse and brain gene expression, Eur J Pharmacol, vol.500, pp.331-349, 2004.

I. Racz, B. Schurmann, A. Karpushova, M. Reuter, S. Cichon et al., The opioid peptides enkephalin and beta-endorphin in alcohol dependence, Biol Psychiatry, vol.64, pp.989-997, 2008.

D. D. Rasmussen, Effects of chronic nicotine treatment and withdrawal on hypothalamic proopiomelanocortin gene expression and neuroendocrine regulation, Psychoneuroendocrinology, vol.23, pp.245-259, 1998.

D. D. Rasmussen, B. M. Boldt, C. A. Bryant, D. R. Mitton, S. A. Larsen et al., Chronic daily ethanol and withdrawal. 1. Long-term changes in the hypothalamo-pituitary-adrenal axis, Alcohol Clin Exp Res, vol.24, pp.1836-1849, 2000.

D. D. Rasmussen, B. M. Boldt, C. W. Wilkinson, and D. R. Mitton, Chronic daily ethanol and withdrawal. 3. Forebrain pro-opiomelanocortin gene expression and implications for dependence, relapse, and deprivation effect, Alcohol Clin Exp Res, vol.26, pp.535-546, 2002.

K. Rasmussen, The role of the locus coeruleus and N-methyl-d-aspartic acid (NMDA) and AMPA receptors in opiate withdrawal, Neuropsychopharmacology, vol.13, pp.295-300, 1995.

S. Rassnick, L. Stinus, and G. F. Koob, The effects of 6-hydroxydopamine lesions of the nucleus accumbens and the mesolimbic dopamine system on oral self-administration of ethanol in the rat, Brain Res, vol.623, pp.16-24, 1993.

W. Renthal and E. J. Nestler, Epigenetic mechanisms in drug addiction, Trends Mol Med, vol.14, pp.341-350, 2008.

A. Rezayof, M. R. Zarrindast, H. Sahraei, and A. H. Haeri-rohani, Involvement of dopamine D2 receptors of the central amygdala on the acquisition and expression of morphine-induced place preference in rat, Pharmacol Biochem Behav, vol.74, pp.187-197, 2002.

J. S. Rhodes and J. C. Crabbe, Gene expression induced by drugs of abuse, Curr Opin Pharmacol, vol.5, pp.26-33, 2005.

P. Y. Risold, R. H. Thompson, and L. W. Swanson, The structural organization of connections between hypothalamus and cerebral cortex, Brain Res, vol.24, pp.197-254, 1997.

T. W. Robbins, M. Cador, J. R. Taylor, and B. J. Everitt, Limbic-striatal interactions in reward-related processes, Neurosci Biobehav Rev, vol.13, pp.155-162, 1989.

A. J. Roberts, L. H. Gold, I. Polis, J. S. Mcdonald, D. Filliol et al., Increased ethanol self-administration in delta-opioid receptor knockout mice, Alcohol Clin Exp Res, vol.25, pp.1249-1256, 2001.

A. J. Roberts, J. S. Mcdonald, C. J. Heyser, B. L. Kieffer, H. W. Matthes et al., mu-Opioid receptor knockout mice do not self-administer alcohol, J Pharmacol Exp Ther, vol.293, pp.1002-1008, 2000.

P. Robledo, V. Mendizabal, J. Ortuno, R. De-la-torre, B. L. Kieffer et al., The rewarding properties of MDMA are preserved in mice lacking mu-opioid receptors, Eur J Neurosci, vol.20, pp.853-858, 2004.

P. Romualdi, A. Donatini, A. Capobianco, and S. Ferri, Methamphetamine alters prodynorphin gene expression and dynorphin A levels in rat hypothalamus, Eur J Pharmacol, vol.365, pp.183-186, 1999.

P. Romualdi, A. Donatini, S. Izenwasser, B. M. Cox, and S. Ferri, Chronic intracerebroventricular cocaine differentially affects prodynorphin gene expression in rat hypothalamus and caudate-putamen, Brain Res, vol.40, pp.153-156, 1996.

P. Romualdi, G. Lesa, and S. Ferri, Chronic opiate agonists downregulate prodynorphin gene expression in rat brain, Brain Res, vol.563, pp.132-136, 1991.

O. K. Ronnekleiv, M. A. Bosch, M. J. Cunningham, E. J. Wagner, D. K. Grandy et al., Downregulation of mu-opioid receptor mRNA in the mediobasal hypothalamus of the female guinea pig following morphine treatment, Neurosci Lett, vol.216, pp.129-132, 1996.

A. Rosin, S. Lindholm, J. Franck, and J. Georgieva, Downregulation of kappa opioid receptor mRNA levels by chronic ethanol and repetitive cocaine in rat ventral tegmentum and nucleus accumbens, Neurosci Lett, vol.275, pp.1-4, 1999.

I. Roth-deri, T. Green-sadan, and G. Yadid, Beta-endorphin and drug-induced reward and reinforcement, Prog Neurobiol, vol.86, pp.1-21, 2008.

I. Roth-deri, R. Mayan, and G. Yadid, A hypothalamic endorphinic lesion attenuates acquisition of cocaine self-administration in the rat, Eur Neuropsychopharmacol, vol.16, pp.25-32, 2006.

J. D. Salamone, M. Correa, A. Farrar, and S. M. Mingote, Effort-related functions of nucleus accumbens dopamine and associated fore-brain circuits, Psychopharmacology, vol.191, pp.461-482, 2007.

J. A. Salinas and J. L. Mcgaugh, The amygdala modulates memory for changes in reward magnitude: involvement of the amygdaloid GABAergic system, Behav Brain Res, vol.80, pp.87-98, 1996.

P. Sanchez-cardoso, A. Higuera-matas, S. Martin, N. Del-olmo, M. Miguens et al., Modulation of the endogenous opioid system after morphine self-administration and during its extinction: a study in Lewis and Fischer 344 rats, Neuropharmacology, vol.52, pp.931-948, 2007.

C. Sanchis-segura and R. Spanagel, Behavioural assessment of drug reinforcement and addictive features in rodents: an overview, Addict Biol, vol.11, pp.2-38, 2006.

A. B. Sante, M. J. Nobre, and M. L. Brandao, Place aversion induced by blockade of mu or activation of kappa opioid receptors in the dorsal periaqueductal gray matter, Behav Pharmacol, vol.11, pp.583-589, 2000.

G. Scherrer, P. Tryoen-toth, D. Filliol, A. Matifas, D. Laustriat et al., Knockin mice expressing fluorescent delta-opioid receptors uncover G protein-coupled receptor dynamics in vivo, Proc Natl Acad Sci USA, vol.103, pp.9691-9696, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00188131

C. W. Schindler, L. V. Panlilio, and S. R. Goldberg, Second-order schedules of drug self-administration in animals, Psychopharmacology, vol.163, pp.327-344, 2002.

W. Schultz, Behavioral dopamine signals, Trends Neurosci, vol.30, pp.203-210, 2007.

Y. Shaham, U. Shalev, L. Lu, D. Wit, H. Stewart et al., The reinstatement model of drug relapse: history, methodology and major findings, Psychopharmacology, vol.168, pp.3-20, 2003.

M. Sharifzadeh, A. Haghighat, P. Tahsili-fahadan, S. Khalaj, M. R. Zarrindast et al., Intrahippocampal inhibition of protein kinase AII attenuates morphine-induced conditioned place preference, Pharmacol Biochem Behav, vol.85, pp.705-712, 2006.

T. P. Sheehan, R. A. Chambers, and D. S. Russell, Regulation of affect by the lateral septum: implications for neuropsychiatry, Brain Res, vol.46, pp.71-117, 2004.

J. Shen, K. W. Chan, B. T. Chen, P. J. Sehba, F. Duttaroy et al., The effect of in vivo ethanol consumption on cyclic AMP and delta-opioid receptors in mouse striatum, Brain Res, vol.770, pp.65-71, 1997.

T. Shimura, H. Imaoka, and T. Yamamoto, Neurochemical modulation of ingestive behavior in the ventral pallidum, Eur J Neurosci, vol.23, pp.1596-1604, 2006.

T. S. Shippenberg and G. I. Elmer, The neurobiology of opiate reinforcement, Crit Rev Neurobiol, vol.12, pp.267-303, 1998.

Y. Shirayama, H. Ishida, M. Iwata, G. I. Hazama, R. Kawahara et al., Stress increases dynorphin immunoreactivity in limbic brain regions and dynorphin antagonism produces antidepressant-like effects, J Neurochem, vol.90, pp.1258-1268, 2004.

J. R. Shoblock and N. T. Maidment, Enkephalin release promotes homeostatic increases in constitutively active mu opioid receptors during morphine withdrawal, Neuroscience, vol.149, pp.642-649, 2007.

R. B. Simerly, Wired for reproduction: organization and development of sexually dimorphic circuits in the mammalian forebrain, Annu Rev Neurosci, vol.25, pp.507-536, 2002.

F. Simonin, O. Valverde, C. Smadja, S. Slowe, I. Kitchen et al., Disruption of the kappa-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective kappaagonist U-50,488H and attenuates morphine withdrawal, EMBO J, vol.17, pp.886-897, 1998.

P. D. Skoubis, H. A. Lam, J. Shoblock, S. Narayanan, and N. T. Maidment, Endogenous enkephalins, not endorphins, modulate basal hedonic state in mice, Eur J Neurosci, vol.21, pp.1379-1384, 2005.

P. D. Skoubis and N. T. Maidment, Blockade of ventral pallidal opioid receptors induces a conditioned place aversion and attenuates acquisition of cocaine place preference in the rat, Neuroscience, vol.119, pp.241-249, 2003.

S. J. Slowe, F. Simonin, B. Kieffer, and I. Kitchen, Quantitative autoradiography of mu-, delta-and kappa1 opioid receptors in kappa-opioid receptor knockout mice, Brain Res, vol.818, pp.335-345, 1999.

K. S. Smith and K. C. Berridge, The ventral pallidum and hedonic reward: neurochemical maps of sucrose "liking" and food intake, J Neurosci, vol.25, pp.8637-8649, 2005.

R. J. Smith, A. , and G. , Noradrenergic transmission in the extended amygdala: role in increased drug-seeking and relapse during protracted drug abstinence, Brain Struct Funct, vol.213, pp.43-61, 2008.

A. R. Soderman and E. M. Unterwald, Cocaine reward and hyperactivity in the rat: sites of mu opioid receptor modulation, Neuroscience, vol.154, pp.1506-1516, 2008.

I. Sora, G. Elmer, M. Funada, J. Pieper, X. F. Li et al., Mu opiate receptor gene dose effects on different morphine actions: evidence for differential in vivo mu receptor reserve, Neuropsychopharmacology, vol.25, pp.41-54, 2001.

R. Spanagel, A. Herz, and T. S. Shippenberg, Opposing tonically active endogenous opioid systems modulate the mesolimbic dopaminergic pathway, Proc Natl Acad Sci USA, vol.89, pp.2046-2050, 1992.

R. Spangler, A. Ho, Y. Zhou, C. E. Maggos, V. Yuferov et al., Regulation of kappa opioid receptor mRNA in the rat brain by "binge" pattern cocaine administration and correlation with pre-prodynorphin mRNA, Brain Res, vol.38, pp.71-76, 1996.

R. Spangler, E. M. Unterwald, and M. J. Kreek, Binge" cocaine administration induces a sustained increase of prodynorphin mRNA in rat caudate-putamen, Brain Res, vol.19, pp.323-327, 1993.

R. Spangler, Y. Zhou, C. E. Maggos, S. D. Schlussman, A. Ho et al., Prodynorphin, proenkephalin and kappa opioid receptor mRNA responses to acute "binge" cocaine, Brain Res, vol.44, pp.139-142, 1997.

B. G. Stanley, D. Lanthier, and S. F. Leibowitz, Multiple brain sites sensitive to feeding stimulation by opioid agonists: a cannula-mapping study, Pharmacol Biochem Behav, vol.31, pp.825-832, 1988.

G. B. Stefano, E. Bianchi, M. Guarna, G. L. Fricchione, W. Zhu et al., Nicotine, alcohol and cocaine coupling to reward processes via endogenous morphine signaling: the dopamine-morphine hypothesis, Med Sci Monit, vol.13, pp.91-102, 2007.

E. A. Stein and J. Olds, Direct intracerebral self-administration of opiates in the rat, Soc Neurosci Abstr, p.302, 1977.

E. A. Stein and J. Zerneskie, Is reward behavior mediated by an endogenous opiate system?, Soc Neurosci Asbstr, p.573, 1979.

H. Steiner and C. R. Gerfen, Cocaine-induced c-fos messenger RNA is inversely related to dynorphin expression in striatum, J Neurosci, vol.13, pp.5066-5081, 1993.

D. N. Stephens, T. Duka, and . Review, Cognitive and emotional consequences of binge drinking: role of amygdala and prefrontal cortex, Philos Trans R Soc Lond B Biol Sci, vol.363, pp.3169-3179, 2008.

L. Stinus, L. Moal, M. Koob, and G. F. , Nucleus accumbens and amygdala are possible substrates for the aversive stimulus effects of opiate withdrawal, Neuroscience, vol.37, pp.767-773, 1990.

I. P. Stolerman, Components of drug dependence: reinforcement, discrimination and adaptation, Biochem Soc Symp, vol.59, pp.1-12, 1993.

P. Svensson and Y. L. Hurd, Specific reductions of striatal prodynorphin and D1 dopamine receptor messenger RNAs during cocaine abstinence, Brain Res, vol.56, pp.162-168, 1998.

L. W. Swanson, Cerebral hemisphere regulation of motivated behavior, Brain Res, vol.886, pp.113-164, 2000.

D. C. Sweet, A. S. Levine, and C. M. Kotz, Functional opioid pathways are necessary for hypocretin-1 (orexin-A)-induced feeding, Peptides, vol.25, pp.307-314, 2004.

S. Tanaka, L. W. Fan, L. T. Tien, Y. Park, L. Y. Liu-chen et al., Butorphanol dependence increases hippocampal kappa-opioid receptor gene expression, J Neurosci Res, vol.82, pp.255-263, 2005.

X. C. Tang, K. Mcfarland, S. Cagle, and P. W. Kalivas, Cocaine-induced reinstatement requires endogenous stimulation of mu-opioid receptors in the ventral pallidum, J Neurosci, vol.25, pp.4512-4520, 2005.

E. Teodorov, C. C. Modena, M. H. Sukikara, and L. F. Felicio, Preliminary study of the effects of morphine treatment on opioid receptor gene expression in brain structures of the female rat, Neuroscience, vol.141, pp.1225-1231, 2006.

M. Terashvili, H. E. Wu, R. J. Leitermann, K. C. Hung, A. D. Clithero et al., Differential conditioned place preference responses to endomorphin-1 and endomorphin-2 microinjected into the posterior nucleus accumbens shell and ventral tegmental area in the rat, J Pharmacol Exp Ther, vol.309, pp.816-824, 2004.

M. Terashvili, H. E. Wu, E. T. Schwasinger, K. C. Hung, J. S. Hong et al., +)-Morphine attenuates the (?)-morphine-produced conditioned place preference and the mu-opioid receptor-mediated dopamine increase in the posterior nucleus accumbens of the rat, Eur J Pharmacol, vol.587, pp.147-154, 2008.

A. J. Tindell, K. S. Smith, S. Pecina, K. C. Berridge, and J. W. Aldridge, Ventral pallidum firing codes hedonic reward: when a bad taste turns good, J Neurophysiol, vol.96, pp.2399-2409, 2006.

G. H. Tjon, P. Voorn, L. J. Vanderschuren, T. J. De-vries, N. H. Michiels et al., Delayed occurrence of enhanced striatal preprodynorphin gene expression in behaviorally sensitized rats: differential long-term effects of intermittent and chronic morphine administration, Neuroscience, vol.76, pp.167-176, 1997.

A. L. Tracy, L. E. Jarrard, and T. L. Davidson, The hippocampus and motivation revisited: appetite and activity, Behav Brain Res, vol.127, pp.13-23, 2001.

S. P. Travers and R. Norgren, Organization of orosensory responses in the nucleus of the solitary tract of rat, J Neurophysiol, vol.73, pp.2144-2162, 1995.

J. Turchan, W. Lason, B. Budziszewska, and B. Przewlocka, Effects of single and repeated morphine administration on the prodynorphin, proenkephalin and dopamine D2 receptor gene expression in the mouse brain, Neuropeptides, vol.31, pp.24-28, 1997.

J. Turchan, M. Maj, B. Przewlocka, and R. Przewlocki, Effect of cocaine and amphetamine on biosynthesis of proenkephalin and prodynorphin in some regions of the rat limbic system, Pol J Pharmacol, vol.54, pp.367-372, 2002.

J. Turchan, B. Przewlocka, W. Lason, and R. Przewlocki, Effects of repeated psychostimulant administration on the prodynorphin system activity and kappa opioid receptor density in the rat brain, Neuroscience, vol.85, pp.1051-1059, 1998.

T. M. Tzschentke, Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade, Addict Biol, vol.12, pp.227-462, 2007.

G. R. Uhl, T. Drgon, C. Johnson, C. Y. Li, C. Contoreggi et al., Molecular genetics of addiction and related heritable phenotypes: genome-wide association approaches identify "connectivity constellation" and drug target genes with pleiotropic effects, Ann NY Acad Sci, vol.1141, pp.318-381, 2008.

G. R. Uhl, J. P. Ryan, and J. P. Schwartz, Morphine alters preproenkephalin gene expression, Brain Res, vol.459, pp.391-397, 1988.

E. M. Unterwald, M. J. Kreek, and M. Cuntapay, The frequency of cocaine administration impacts cocaine-induced receptor alterations, Brain Res, vol.900, pp.103-109, 2001.

F. J. Vaccarino, H. O. Pettit, F. E. Bloom, and G. F. Koob, Effects of intra-cerebroventricular administration of methyl naloxonium chloride on heroin self-administration in the rat, Pharmacol Biochem Behav, vol.23, pp.495-498, 1985.

F. J. Vaccarino and M. R. Taube, Intra-arcuate opiate actions stimulate GRF-dependent and proteinselective feeding, Peptides, vol.18, pp.197-205, 1997.

E. J. Van-bockstaele, J. Peoples, A. S. Menko, K. Mchugh, and G. Drolet, Decreases in endogenous opioid peptides in the rat medullocoerulear pathway after chronic morphine treatment, J Neurosci, vol.20, pp.8659-8666, 2000.

W. R. Van-furth, M. G. Van-emst, and J. M. Van-ree, Opioids and sexual behavior of male rats: involvement of the medial preoptic area, Behav Neurosci, vol.109, pp.123-134, 1995.

W. R. Van-furth and J. M. Van-ree, Sexual motivation: involvement of endogenous opioids in the ventral tegmental area, Brain Res, vol.729, pp.20-28, 1996.

W. R. Van-furth, G. Wolterink, and J. M. Van-ree, Regulation of masculine sexual behavior: involvement of brain opioids and dopamine, Brain Res, vol.21, pp.162-184, 1995.

J. M. Van-ree and D. De-wied, Involvement of neurohypophyseal peptides in drug-mediated adaptive responses, Pharmacol Biochem Behav, vol.13, issue.1, pp.257-263, 1980.

J. M. Van-ree, M. A. Gerrits, and L. J. Vanderschuren, Opioids, reward and addiction: an encounter of biology, psychology, and medicine, Pharmacol Rev, vol.51, pp.341-396, 1999.

J. M. Van-ree, R. J. Niesink, L. Van-wolfswinkel, N. F. Ramsey, M. M. Kornet et al., Van den Berg CL. Endogenous opioids and reward, Eur J Pharmacol, vol.405, pp.89-101, 2000.

J. M. Van-ree and N. Ramsey, The dopamine hypothesis of opiate reward challenged, Eur J Pharmacol, vol.134, pp.239-243, 1987.

L. J. Vanderschuren and B. J. Everitt, Drug seeking becomes compulsive after prolonged cocaine selfadministration, Science, vol.305, pp.1017-1019, 2004.

A. Vecchiola, P. Collyer, R. Figueroa, R. Labarca, G. Bustos et al., Differential regulation of mu-opioid receptor mRNA in the nucleus accumbens shell and core accompanying amphetamine behavioral sensitization, Brain Res, vol.69, pp.1-9, 1999.

N. D. Volkow and T. K. Li, Drugs and alcohol: treating and preventing abuse, addiction and their medical consequences, Pharmacol Ther, vol.108, pp.3-17, 2005.

B. M. Walker and A. Ettenberg, Intra-ventral tegmental area heroin-induced place preferences in rats are potentiated by peripherally administered alprazolam, Pharmacol Biochem Behav, vol.82, pp.470-477, 2005.

J. R. Walker, S. H. Ahmed, K. N. Gracy, and G. F. Koob, Microinjections of an opiate receptor antagonist into the bed nucleus of the stria terminalis suppress heroin self-administration in dependent rats, Brain Res, vol.854, pp.85-92, 2000.

C. L. Walters, J. N. Cleck, Y. C. Kuo, and J. A. Blendy, Mu-opioid receptor and CREB activation are required for nicotine reward, Neuron, vol.46, pp.933-943, 2005.

H. Wang and M. W. Wessendorf, Mu-and delta-opioid receptor mRNAs are expressed in periaqueductal gray neurons projecting to the rostral ventromedial medulla, Neuroscience, vol.109, pp.619-634, 2002.

M. A. Waraczynski, The central extended amygdala network as a proposed circuit underlying reward valuation, Neurosci Biobehav Rev, vol.30, pp.472-496, 2006.

H. G. Ward, D. M. Nicklous, V. J. Aloyo, and K. J. Simansky, Mu-opioid receptor cellular function in the nucleus accumbens is essential for hedonically driven eating, Eur J Neurosci, vol.23, pp.1605-1613, 2006.

H. G. Ward and K. J. Simansky, Chronic prevention of mu-opioid receptor (MOR) G-protein coupling in the pontine parabrachial nucleus persistently decreases consumption of standard but not palatable food, Psychopharmacology, vol.187, pp.435-446, 2006.

S. J. Ward, T. J. Martin, and D. C. Roberts, Beta-funaltrexamine affects cocaine self-administration in rats responding on a progressive ratio schedule of reinforcement, Pharmacol Biochem Behav, vol.75, pp.301-307, 2003.

S. J. Ward and D. C. Roberts, Microinjection of the delta-opioid receptor selective antagonist naltrindole 5?-isothiocyanate site specifically affects cocaine self-administration in rats responding under a progressive ratio schedule of reinforcement, Behav Brain Res, vol.182, pp.140-144, 2007.

S. L. Wardlaw, J. Kim, and S. Sobieszczyk, Effect of morphine on proopiomelanocortin gene expression and peptide levels in the hypothalamus, Brain Res, vol.41, pp.140-147, 1996.

R. Weissenborn, R. B. Whitelaw, T. W. Robbins, and B. J. Everitt, Excitotoxic lesions of the mediodorsal thalamic nucleus attenuate intravenous cocaine self-administration, Psychopharmacology, vol.140, pp.225-232, 1998.

M. Werme, P. Thoren, L. Olson, and S. Brene, Running and cocaine both upregulate dynorphin mRNA in medial caudate putamen, Eur J Neurosci, vol.12, pp.2967-2974, 2000.

M. J. Will, E. B. Franzblau, and A. E. Kelley, The amygdala is critical for opioid-mediated binge eating of fat, Neuroreport, vol.15, pp.1857-1860, 2004.

M. J. Will, W. E. Pratt, and A. E. Kelley, Pharmacological characterization of high-fat feeding induced by opioid stimulation of the ventral striatum, Physiol Behav, vol.89, pp.226-234, 2006.

J. T. Williams, M. J. Christie, and O. Manzoni, Cellular and synaptic adaptations mediating opioid dependence, Physiol Rev, vol.81, pp.299-343, 2001.

J. D. Wilson, D. M. Nicklous, V. J. Aloyo, and K. J. Simansky, An orexigenic role for mu-opioid receptors in the lateral parabrachial nucleus, Am J Physiol Regul Integr Comp Physiol, vol.285, pp.1055-1065, 2003.

A. Winkler, B. Buzas, W. E. Siems, G. Heder, and B. M. Cox, Effect of ethanol drinking on the gene expression of opioid receptors, enkephalinase, and angiotensin-converting enzyme in two inbred mice strains, Alcohol Clin Exp Res, vol.22, pp.1262-1271, 1998.

R. A. Wise and . Dopamine, learning and motivation, Nat Rev Neurosci, vol.5, pp.483-494, 2004.

J. D. Woolley, B. S. Lee, and H. L. Fields, Nucleus accumbens opioids regulate flavor-based preferences in food consumption, Neuroscience, vol.143, pp.309-317, 2006.

J. D. Woolley, B. S. Lee, B. Kim, and H. L. Fields, Opposing effects of intra-nucleus accumbens mu and kappa opioid agonists on sensory specific satiety, Neuroscience, vol.146, pp.1445-1452, 2007.

J. D. Woolley, B. S. Lee, S. A. Taha, and H. L. Fields, Nucleus accumbens opioid signaling conditions shortterm flavor preferences, Neuroscience, vol.146, pp.19-30, 2007.

R. C. Wright and A. J. Ingenito, Blockade of dorsal hippocampal kappa-opioid receptors increases blood pressure in normotensive and isolation-induced hypertensive rats, Neuropeptides, vol.37, pp.127-132, 2003.

F. Yan and B. L. Roth, Salvinorin A: a novel and highly selective kappa-opioid receptor agonist, Life Sci, vol.75, pp.2615-2619, 2004.

L. J. Young, M. M. Lim, B. Gingrich, and T. R. Insel, Cellular mechanisms of social attachment, Horm Behav, vol.40, pp.133-138, 2001.

V. Yuferov, D. Nielsen, E. Butelman, and M. J. Kreek, Microarray studies of psychostimulant-induced changes in gene expression, Addict Biol, vol.10, pp.101-118, 2005.

V. Yuferov, Y. Zhou, K. S. Laforge, R. Spangler, A. Ho et al., Elevation of guinea pig brain preprodynorphin mRNA expression and hypothalamic-pituitary-adrenal axis activity by "binge" pattern cocaine administration, Brain Res Bull, vol.55, pp.65-70, 2001.

D. S. Zahm, L. Zaborszky, V. E. Alones, and L. Heimer, Evidence for the coexistence of glutamate decarboxylase and Met-enkephalin immunoreactivities in axon terminals of rat ventral pallidum, Brain Res, vol.325, pp.317-321, 1985.

A. Zangen, S. Ikemoto, J. E. Zadina, and R. A. Wise, Rewarding and psychomotor stimulant effects of endomorphin-1: anteroposterior differences within the ventral tegmental area and lack of effect in nucleus accumbens, J Neurosci, vol.22, pp.7225-7233, 2002.

M. R. Zarrindast, S. Ahmadi, A. Haeri-rohani, A. Rezayof, M. R. Jafari et al., A) receptors in the basolateral amygdala are involved in mediating morphine reward, Brain Res, vol.1006, pp.49-58, 2004.

M. R. Zarrindast, M. Ebrahimi-ghiri, P. Rostami, and A. Rezayof, Repeated pre-exposure to morphine into the ventral pallidum enhances morphine-induced place preference: involvement of dopaminergic and opioidergic mechanisms, Behav Brain Res, vol.181, pp.35-41, 2007.

H. Zheng, L. M. Patterson, and H. R. Berthoud, Orexin signaling in the ventral tegmental area is required for high-fat appetite induced by opioid stimulation of the nucleus accumbens, J Neurosci, vol.27, pp.11075-11082, 2007.

Y. Zhou, J. Bendor, L. Hofmann, M. Randesi, A. Ho et al., Mu opioid receptor and orexin/ hypocretin mRNA levels in the lateral hypothalamus and striatum are enhanced by morphine withdrawal, J Endocrinol, vol.191, pp.137-145, 2006.

Y. Zhou, J. T. Bendor, V. Yuferov, S. D. Schlussman, A. Ho et al., Amygdalar vasopressin mRNA increases in acute cocaine withdrawal: evidence for opioid receptor modulation, Neuroscience, vol.134, pp.1391-1397, 2005.

Y. Zhou, C. L. Cui, S. D. Schlussman, J. C. Choi, A. Ho et al., Effects of cocaine place conditioning, chronic escalating-dose "binge" pattern cocaine administration and acute withdrawal on orexin/hypocretin and preprodynorphin gene expressions in lateral hypothalamus of Fischer and Sprague-Dawley rats, Neuroscience, vol.153, pp.1225-1234, 2008.

B. Ziolkowska, R. Stefanski, P. Mierzejewski, G. Zapart, W. Kostowski et al., Contingency does not contribute to the effects of cocaine self-administration on prodynorphin and proenkephalin gene expression in the rat forebrain, Brain Res, vol.1069, pp.1-9, 2006.

C. Zollner, C. Stein, and . Opioids, Bottom panel maps cell bodies expressing opioid peptides, as evaluated both by immunohistochemical and in situ hybridization studies. Note: for immunohistochemical mapping, data based on antibodies for peptide precursors were used in priority. When not available, data based on antibodies for final peptides were used, with priority given to peptides issued from a single precursor (?-endorphin and dynorphin), Handb Exp Pharmacol, pp.31-63, 2007.

, CeA, central nucleus, amygdala; Cl, claustrum; CL, centrolateral thalamus; CM

. Coa, . Cpu, and . Crbn,

. Dmh,

;. Dmr and . Dtn,

, En, endopiriform cortex

. Ent,

G. and N. Gelatinosus,

G. Vp, globus pallidus/ventral pallidum; HbL, lateral habenula

. Hbm,

, HPC, hippocampus; IL, intermediate lobe, pituitary; IP

L. Lc and . Coeruleus,

, LRN, lateral reticular nucleus; MD, mediodorsal thalamus; Me, median eminence

, MEA, median nucleus, amygdala; MG, medial geniculate; MM, medial mammillary nucleus

, MV, medial vestibular nucleus; NAc, nucleus accumbens; NL, neuronal lobe, pituitary; NRGC, nucleus reticularis gigantocellularis; NTS, nucleus tractus solitarius

. Ocx, . Pag, and . Gray, PCx, parietal cortex; Pir, piriform cortex; PN, pontine nucleus; PnR, pontine reticular; PO, posterior thalamus; POA, preoptic area

. Prs and . Pv, PVN, paraventricular hypothalamus; RE, reuniens thalamus; RN, red nucleus; RM, raphé magnus; SON, supraoptic nucleus; SN, substancia nigra; SNT, sensory trigeminal nucleus

, STN, spinal trigeminal nucleus

. Tcx,

. Vm and . Vmh,

. Vpl and . Vta,

Z. Zi and . Incerta,

. Pvn and . Vp,

, VTA, ventral tegmental area. express all three types of opioid receptors. Amb, nucleus ambiguus

. Amg,

, BNST, bed nucleus of the stria terminalis

. Cpu,

. Hpc and . Lh,

. Pag and . Gray,

, POA, preoptic area; PPTg, pedunculopontine nucleus

R. and R. Nucleus,

S. Sn and ;. Vp,

, MM, medial mammillary nucleus

M. Mo and . Oblongata,

. Nac, accumbens; NL, neuronal lobe, pituitary; PAG, nucleus

. Pgi,

. Vmh,