<<

6.- BIBLIOGRAFÍA BIBLIOGRAFÍA

Abbott F.V., Franklin K.B., Westbrook R.F. The formalin test: scoring properties of the first and second phases of the pain response in rats. Pain. (1995). 60: 91- 102.

Abdelhamid E.E., Sultana M., Portoghese P.S., Takemori A.E. Selective blockage of delta receptors prevents the development of tolerance and dependence in mice. J Pharmacol Exp Ther. (1991). 258: 299-303.

Acosta C.G., Lopez H.S. Delta modulation of several voltage- dependent Ca(2+) currents in rat sensory neurons. J Neurosci. (1999). 19: 8337-8348.

Adams, J.U., Tallarida, R.J., Geller, E.B., Adler, M.W. Isobolographic Superadditivity detween Delta and Mu Opioid in the Rat Depends on the Ratio of Compounds, the Mu and the Assay Used. J Pharmacol Exp Ther. (1993). 266: 1261-1267.

Aghajanian G.K., Wang Y.Y. Pertussis toxin blocks the outward currents evoked by and alpha 2-agonists in locus coeruleus neurons. Brain Res. (1986). 371: 390-394.

Akil H., Madden J., Patrick R.L., Barchas J.D. Stress-induced increase in endogenous opiate peptides; concurrent analgesia and its partial reversal by . En Koterlitz H.W. (Eds.). and Endogenous Opioid Peptides, Elsevier, Amsterdam. (1976). 63-70.

Akiyama Y., Nishimura M., Suzuki A., Yamamoto M., Kishi F., Kawakami Y. Naloxone increases ventilatory response to hypercapnic hypoxia in healthy adult humans. Am Rev Respir Dis. (1990). 142: 301-305.

Albrecht E., Heinrich N., Lorenz D., Baeger I., Samovilova N., Fechner K., Berger H. Influence of continuous levels of in rats on the mu-opioid receptor in the central nervous system. Pharmacol Biochem Behav. (1997). 58: 189-194.

203 BIBLIOGRAFÍA

Alvan G., Bechtel P., Iselius L., Gundert-Remy U. Hydroxylation polymorphisms of debrisoquine and mephenytoin in European populations. Eur J Clin Pharmacol. (1990). 39: 533-537.

American Psychiatric Association. Diagnostics and statistical manual of mental disorders. 4th Edition (1994). Washinton DC, American Psychiatric Press.

Anagnostakis Y., Zis V., Spyraki C. Analgesia induced by morphine injected into the pallidum. Behav Brain Res. (1992). 48: 135-143.

Angers, S., Salahpour, A., Bouvier, M. Biochemical and biophysical demonstration of GPCR oligomerization in mammalian cells. Life Sci. (2001). 68: 2243-2250.

Antunes Bras J.M., Laporte A.M., Benoliel J.J., Bourgoin S., Mauborgne A., Hamon M., Cesselin F., Pohl M. Effects of peripheral axotomy on cholecystokinin neurotransmission in the rat spinal cord. J Neurochem. (1999). 72: 858-867.

Appelbaum B.D., Holtzman S.G. Restraint stress enhances morphine-induced analgesia in the rat without changing apparent affinity of receptor. Life Sci. (1985). 36: 1069-1074.

Attali B., Gouarderes C., Mazarguil H., Audigier Y., Cros J. Evidence for multiple "Kappa" binding sites by use of opioid peptides in the guinea-pig lumbo-sacral spinal cord. Neuropeptides. (1982). 3: 53-64.

Atweh S.F., Kuhar M.J. Distribution and physiological significance of opioide receptors in the brain. British Medical Bulletin. (1983) 39: 47-52.

Avidor-Reiss T., Nevo I., Levy R., Pfeuffer T., Vogel Z. Chronic opioid treatment induces adenylyl cyclase V superactivation. Involvement of Gbetagamma. J Biol Chem. (1996). 271: 21309-21315.

Baker M.L., Brockunier L.L., Bagley J.R., France C.P., Carr D.J. Fentanyl-related 4- heteroanilido piperidine OHM3295 augments splenic natural killer activity and

204 BIBLIOGRAFÍA

induces analgesia through opioid receptor pathways. J Pharmacol Exp Ther. (1995). 274: 1285-1292.

Baker A.K., Hoffmann V.L., Meert T.F. and potentiate the antinociceptive effects of mu- but not delta- or kappa-opioid agonists in a mouse model of acute pain. Pharmacol Biochem Behav. (2002). 74: 73-86.

Baker L., Ratka A. Sex-specific differences in levels of morphine, morphine-3- glucuronide, and morphine antinociception in rats. Pain. (2002). 95: 65-74.

Ballet S., Braz J., Mauborgne A., Bourgoin S., Zajac J.M., Hamon M., Cesselin F. The neuropeptide FF analogue, 1DMe, reduces in vivo release from the rat spinal cord. J Neurochem. (2002). 81: 659-662.

Ballet S., Mauborgne A., Gouarderes C., Bourgoin A.S., Zajac J.M., Hamon M., Cesselin F. The neuropeptide FF analogue, 1DME, enhances in vivo met- release from the rat spinal cord. Neuropharmacology. (1999). 38: 1317-1324.

Bals-Kubik R., Ableitner A., Herz A., Shippenberg T.S. Neuroanatomical sites mediating the motivational effects of as mapped by the conditioned place preference paradigm in rats. J Pharmacol Exp Ther. (1993). 264: 489- 495.

Baran A., Shuster L., Eleftheriou B.E., Bailey D.W. Opiate receptors in mice: genetic differences. Life Sci. (1975). 17: 633-640.

Basbaum A.I. y Fields H.L. Endogenous pain control systems: brainstem spinal pathways and endorphin circuitry. Annu Rev Neurosci. (1984). 7: 309-338.

Baumeister A.A., Hawkins M.F., Anticich T.G., Moore L.L., Higgins T.D. Bilateral intranigral microinjection of morphine and opioid peptides produces antinociception in rats. Brain Res. (1987). 411: 183-6.

205 BIBLIOGRAFÍA

Beecher H.K. Measurement of subjetive responses, Oxford University Press, New York. (1959).

Belcheva M.M., Vogel Z., Ignatova E., Avidor-Reiss T., Zippel R., Levy R., Young E.C., Barg J., Coscia C.J. Opioid modulation of extracellular signal-regulated protein kinase activity is ras-dependent and involves Gbetagamma subunits. J Neurochem. (1998). 70: 635-645.

Belknap J.K., Danielson P.W., Laursen S.E., Noordewier B. Selective breeding for -induced antinociception on the hot-plate assay: commonalities in mechanism of action with morphine, , ethylketocyclazocine, U- 50488H and clonidine in mice. J Pharmacol Exp Ther. (1987). 241: 477-481.

Bennett G.J. Animal models of pain. En: Kruger L. (Eds.). Methods in pain research. (2001): 67-91.

Berhow M.T., Hiroi N., Nestler E.J. Regulation of ERK (extracellular signal regulated kinase), part of the neurotrophin signal transduction cascade, in the rat mesolimbic dopamine system by chronic exposure to morphine or cocaine. J Neurosci. (1996). 16: 4707-4715.

Bian D., Ossipov M.H., Ibrahim M., Raffa R.B., Tallarida R.J., Malan T.P. Jr., Lai J., Porreca F. Loss of antiallodynic and antinociceptive spinal/supraspinal morphine synergy in nerve-injured rats: restoration by MK-801 or dynorphin antiserum. Brain Res. (1999). 831: 55-63.

Bilsky E.J., Calderon S.N., Wang T., Bernstein R.N., Davis P., Hruby V.J., McNutt R.W., Rothman R.B., Rice K.C., Porreca F. SNC 80, a selective, nonpeptidic and systemically active opioid delta agonist. J Pharmacol Exp Ther. (1995). 273: 359-366.

Birnbaumer L., Abramowitz J., Brown A.M. Receptor-effector coupling by G proteins. Biochim Biophys Acta. (1990). 1031: 163-224.

206 BIBLIOGRAFÍA

Blume A.J. Opiate binding to membrane preparations of neuroblastoma x glioma hybrid cells NG108-15: effects of ions and nucleotides. Life Sci. (1978). 22: 1843-1852.

Blume A.J., Lichtshtein D., Boone G. Coupling of opiate receptors to adenylate cyclase: requirement for Na+ and GTP. Proc Natl Acad Sci U S A. (1979). 76: 5626-5630.

Bodnar R.J. Supraspinal circuitry mediating opioid antinociception: antagonist and synergy studies in multiple sites. J Biomed Sci. (2000). 7: 181-194.

Bodnar R.J., Hadjimarkou M.M. Endogenous opiates and behavior: 2002. Peptides. (2003). 24: 1241-1302.

Bolan E.A., Tallarida R.J., Pasternak G.W. Synergy between mu opioid ligands: evidence for functional interactions among mu opioid receptor subtypes. J Pharmacol Exp Ther. (2002). 303: 557-562.

Bonner G., Meng F., Akil H. Selectivity of mu-opioid receptor determined by interfacial residues near third extracellular loop. Eur J Pharmacol. (2000). 403:37-44.

Bourgoin S., Le Bars D., Clot A.M., Hamon M., Cesselin F. Subcutaneous formalin induces a segmental release of Met-enkephalin-like material from the rat spinal cord. Pain. (1990). 41: 323-329.

Bozarth M.A., Wise R.A. Anatomically distinct opiate receptor fields mediate reward and physical dependence. Science. (1984). 224: 516-517.

Brandt M., Gullis R.J., Fischer K., Buchen C., Hamprecht B., Moroder L., Wunsch E. Enkephalin regulates the levels of cyclic nucleotides in neuroblastoma x glioma hybrid cells. Nature. (1976). 262: 311-313.

Bulka A., Kouya P.F., Bottiger Y., Svensson J.O., Xu X.J., Wiesenfeld-Hallin Z. Comparison of the antinociceptive effect of morphine, ,

207 BIBLIOGRAFÍA

and in two substrains of Sprague-Dawley rats. Eur J Pharmacol. (2004). 492: 27-34.

Bulka A., Wiesenfeld-Hallin Z., Xu X.J. Differential antinociception by morphine and methadone in two sub-strains of Sprague-Dawley rats and its potentiation by dextromethorphan. Brain Res. (2002). 942: 95-100.

Butelman E.R., Ko M.C., Sobczyk-Kojiro K., Mosberg H.I., Van Bemmel B., Zernig G., Woods J.H. Kappa-Opioid receptor binding populations in rhesus monkey brain: relationship to an assay of thermal antinociception. J Pharmacol Exp Ther. (1998). 285: 595-601.

Buzas B., Toth G., Cavagnero S., Hruby V.J., Borsodi A. Synthesis and binding characteristics of the highly delta-specific new tritiated , [3H] II. Life Sci. (1992). 50:PL75-8.

Calcagnetti D.J., Fleetwood S.W., Holtzman S.G. Pharmacological profile of the potentiation of opioid analgesia by restraint stress. Pharmacol Biochem Behav. (1990). 37: 193-199.

Camí J., Ayesta F.J. Farmacodependencias. En: Flórez J., Armijo J.A., Mediavilla A. (Eds.). Farmacología humana. 3ª Ed. (1997): 565-606.

Carrasco J.L. Homogeneidad de un conjunto de muestras: Análisis de la varianza para dos factores. En: Carrasco J.L. (Eds). El método estadístico en la investigación médica. (1986): 253-260.

Carter R.B. Differentiating analgesic and non-analgesic drug activities on rat hot plate: effect of behavioral endpoint. Pain. (1991). 47: 211-220.

Castanas E., Giraud P., Bourhim N., Cantau P., Oliver C. Kappa 3: a novel subtype of the kappa opioid site in bovine adrenal medulla, Highly selective for Met- enkephalin-Arg6-Phe7. Neuropeptides. (1984). 5: 133-136.

208 BIBLIOGRAFÍA

Castillo R., Kissin I., Bradley E.L. Selective kappa opioid agonist for spinal analgesia without the risk of respiratory depression. Anesth Analg. (1986). 65: 350-354.

Cha X.Y., Xu H., Ni Q., Partilla J.S., Rice K.C., Matecka D., Calderon S.N., Porreca F., Lai J., Rothman R.B. Opioid peptide receptor studies. 4. Antisense oligodeoxynucleotide to the delta opioid receptor delineates opioid receptor subtypes. Regul Pept. (1995). 59: 247-253.

Chang K.J., Hazum E., Killian A., Cuatrecasas P. Interactions of ligands with morphine and enkephalin receptors are differentially affected by guanine nucleotide. Mol Pharmacol. (1981). 20: 1-7.

Chang K.J., Rigdon G.C., Howard J.L., McNutt R.W. A novel, potent and selective nonpeptidic delta opioid receptor agonist BW373U86. J Pharmacol Exp Ther. (1993). 267: 852-857.

Chen Z.R., Irvine R.J., Somogyi A.A., Bochner F. Mu receptor binding of some commonly used opioids and their metabolites. Life Sci. (1991). 48: 2165-2171.

Chen Y., Mestek A., Liu J., Hurley J.A., Yu L. Molecular cloning and functional expression of a µ-opioid receptor from rat brain. Mol Pharmacol. (1993). 44: 8- 12.

Cheng Y., Prusoff W.H. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol. (1973). 22: 3099-3108.

Chesler E.J., Wilson S.G., Lariviere W.R., Rodriguez-Zas S.L., Mogil J.S. Identification and ranking of genetic and laboratory environment factors influencing a behavioral trait, thermal nociception, via computational analysis of a large data archive. Neurosci Biobehav Rev. (2002). 26: 907-923.

Childers S.R. Opioid receptor-coupled second messenger systems. Life Sci. (1991). 48: 1991-2003.

209 BIBLIOGRAFÍA

Childers S.R, Snyder S.H. Differential regulation by guanine nucleotides or opiate agonist and antagonist receptor interactions. J Neurochem. (1980). 34: 583-593.

Childers S.R., Xiao R., Vogt L., Sim L.J. Lack of evidence of kappa2-selective activation of G-proteins: kappa opioid receptor stimulation of [35S] GTPgammaS binding in guinea pig brain. Biochem Pharmacol. (1998). 56: 113-120.

Ching M. Morphine suppresses the proestrous surge of GnrH in pituitary portal plasma of rats. Endocrinology. (1983). 112: 2209-2211.

Clark M.J., Emmerson P.J., Mansour A., Akil H., Woods J.H., Portoghese P.S., Remmers A.E., Medzihradsky F. Opioid efficacy in a C6 glioma cell line stably expressing the delta opioid receptor. J Pharmacol Exp Ther. (1997). 283: 501- 510.

Clark J.A., Liu L., Price M., Hersh B., Edelson M., Pasternak G.W. Kappa opiate receptor multiplicity: evidence for two U50488-sensitive kappa 1 subtypes and a novel kappa 3 subtype. J Pharmacol Exp Ther. (1989). 251: 461-468.

Clausen T.R., Moller M., Woldbye D.P. Inhibitory effect of neuropeptide Y on morphine withdrawal is accompanied by reduced c-fos expression in specific brain regions. J Neurosci Res. (2001). 64: 410-417.

Cleary J., Mikus G., Somogyi A., Bochner F. The influence of pharmacogenetics on opioid analgesia: studies with codeine and in the Sprague-Dawley/ Dark Agouti rat model. J Pharmacol Exp Ther. (1994). 271: 1528-1534.

Collier H.O., Dinneen L.C., Johnson C.A., Schneider C. The abdominal constriction response and its suppression by analgesic drugs in the mouse. Br J Pharmacol. (1968). 32: 295-310.

Collier H.O., Roy A.C. Morphine-like drugs inhibit the stimulation of E prostaglandins of cyclic AMP formation by rat brain homogenate. Nature. (1974). 248: 24-27.

210 BIBLIOGRAFÍA

Collin E., Poulain P., Gauvain-Piquard A., Petit G., Pichard-Leandri E. Is disease progression the major factor in morphine 'tolerance' in cancer pain treatment? Pain. (1993). 55: 319-326.

Colpaert F.C. System theory of pain and of opiate analgesia: no tolerance to opiates. Pharmacol Rev. (1996). 48: 355-402.

Colpaert F.C., Niemegeers C.J., Janssen P.A., Maroli A.N. The effects of prior fentanyl administration and of pain on fentanyl analgesia: tolerance to and enhancement of analgesia. J Pharmacol Exp Ther. (1980). 213: 418- 424.

Comer S.D., McNutt R.W., Chang K.J., De Costa B.R., Mosberg H.I., Woods J.H. Discriminative stimulus effects of BW373U86: a nonpeptide ligand with selectivity for delta opioid receptors. J Pharmacol Exp Ther. (1993). 267: 866- 874.

Connor M., Christie M.D. Opioid receptor signalling mechanisms. Clin Exp Pharmacol Physiol. (1999). 26: 493-499.

Contreras P.C., Tam L., Drower E., Rafferty M.F. [3H]: a potent and selective ligand for labeling delta-opioid receptors. Brain Res. (1993). 604:160- 164.

Cook C.D., Barrett A.C., Roach E.L., Bowman J.R., Picker M.J. Sex-related differences in the antinociceptive effects of opioids: importance of rat genotype, nociceptive stimulus intensity, and efficacy at the mu opioid receptor. Psychopharmacology. (2000). 150: 430-442.

Cotton R., Kosterlitz H.W., Paterson S.J., Rance M.J., Traynor J.R. The use of [3H]-

[D-Pen2,D-Pen5]enkephalin as a highly selective ligand for the delta-binding site. Br J Pharmacol. (1985). 84: 927-932.

211 BIBLIOGRAFÍA

Couceyro P., Douglass J. Precipitated morphine withdrawal stimulates multiple activator protein-1 signaling pathways in rat brain. Mol Pharmacol. (1995). 47: 29-39.

Cox B.M. Mecanismos de tolerancia. En: Stein C (Eds). Opioides en el control del dolor. Aspectos básicos y clínicos. Masson, Barcelona (2001). 105-125.

Craft R.M., Tseng A.H., McNiel D.M., Furness M.S., Rice K.C. Receptor-selective antagonism of opioid antinociception in female versus male rats. Behav Pharmacol. (2001). 12: 591-602.

Cunningham C.L., Niehus D.R., Malott D.H., Prather L.K. Genetic differences in the rewarding and activating effects of morphine and ethanol. Psychopharmacology. (1992). 107: 385-393.

Cvejic S., Devi L.A. Dimerization of the delta opioid receptor: implication for a role in receptor internalization. J Biol Chem (1997). 272: 26959-26964.

D´Amour F.E., Smith D.L. A method for determining loss of pain sensation. J. Pharmacol. Exp. Ther. (1941). 74: 74-79.

D'Anci K.E., Gerstein A.V., Kanarek R.B. Long-term voluntary access to running wheels decreases kappa-opioid antinociception. Pharmacol Biochem Behav. (2000). 66: 343-346.

Dawson-Basoa M.E., Gintzler A.R. Estrogen and progesterone activate spinal kappa- opiate receptor analgesic mechanisms. Pain. (1996). 64: 608-615.

Dawson-Basoa M., Gintzler A.R. Gestational and ovarian sex steroid antinociception: synergy between spinal kappa and delta opioid systems. Brain Res. (1998). 794: 61-67.

Dayer P., Desmeules J., Leemann T., Striberni R. Bioactivation of the narcotic drug codeine in human liver is mediated by the polymorphic monooxygenase

212 BIBLIOGRAFÍA

catalyzing debrisoquine 4-hydroxylation (cytochrome P-450 dbl/bufI). Biochem Biophys Res Commun. (1988). 152: 411-416.

De Araujo Lucas G., Alster P., Brodin E., Wiesenfeld-Hallin Z. Differential release of cholecystokinin by morphine in rat spinal cord. Neurosci Lett. (1998). 245: 13-16.

De A., Stadel J.M., Lefkowitz R.J. A ternary complex model explains the agonist-specific binding properties of the adenylate cyclase-coupled beta- adrenergic receptor. J Biol Chem. (1980). 255: 7108-7117.

Desmeules J., Gascon M.P., Dayer P., Magistris M. Impact of environmental and genetic factors on codeine analgesia. Eur J Clin Pharmacol. (1991). 41: 23-26.

De Vries T.J., Schoffelmeer A.N., Binnekade R., Mulder A.H., Vanderschuren L.J. Drug-induced reinstatement of - and cocaine-seeking behaviour following long-term extinction is associated with expression of behavioural sensitization. Eur J Neurosci. (1998). 10: 3565-3571.

Dhawan B.N., Cesselin F., Raghubir R., Reisine T., Bradley P.B., Portoghese P.S., Hamon M. International Union of Pharmacology. XII. Classification of opioid receptors. Pharmacol Rev. (1996). 48: 567-592.

Di Chiara G. y North R.A. Neurobiology of opiate abuse. Trends. Pharmacol. Sci. (1992). 13: 185-193.

Dickenson A.H. Mechanisms of the analgesic actions of opiates and opioids. Br Med Bull. (1991). 47: 690-702.

Dickenson A.H., Sullivan A.F. Peripheral origins and central modulation of subcutaneous formalin-induced activity of rat dorsal horn neurones. Neurosci Lett. (1987). 83: 207-211.

213 BIBLIOGRAFÍA

Doupnik C.A., Davidson N., Lester H.A., Kofuji P. RGS proteins reconstitute the rapid gating kinetics of Gβγ-activated inwardly rectifying K+ channels. Proc. Natl. Acad. Sci. USA. (1997). 94: 10461-10466.

Dourish C.T., O'Neill M.F., Coughlan J., Kitchener S.J., Hawley D., Iversen S.D. The selective CCK-B L-365,260 enhances morphine analgesia and prevents morphine tolerance in the rat. Eur J Pharmacol. (1990). 176: 35-44.

Dubinsky B., Gebre-Mariam S., Capetola R.J., Rosenthale M.E. The antialgesic drugs: human therapeutic correlates of their potency in laboratory animal models of hyperalgesia. Agents Actions. (1987). 20: 50-60.

Dubuisson D., Dennis S.G. The formalin test: a quantitative study of the analgesic effects of morphine, meperidine, and brain stem stimulation in rats and cats. Pain. (1977). 4: 161-174.

Duman R.S., Tallman J.F., Nestler E.J. Acute and chronic opiate-regulation of adenylate cyclase in brain: specific effects in locus coeruleus. J Pharmacol Exp Ther. (1988). 246: 1033-1039.

Ebert B., Andersen S., Krogsgaard-Larsen P. , methadone and are non-competitive N-methyl-D-aspartate (NMDA) antagonists in the rat cortex and spinal cord. Neurosci. Lett. (1995). 187: 165-168.

Elliott J., Guo L., Traynor J.R. Tolerance to mu-opioid agonists in human neuroblastoma SH-SY5Y cells as determined by changes in guanosine-5'-O-(3- [35S]-thio)triphosphate binding. Br J Pharmacol. (1997). 121: 1422-1428.

Endoh T., Matsuura H., Tajima A., Izumimoto N., Tajima C., Suzuki T., Saitoh A., Suzuki T., Narita M., Tseng L., Nagase H. Potent antinociceptive effects of TRK-820, a novel kappa-opioid receptor agonist. Life Sci. (1999). 65: 1685- 1694

214 BIBLIOGRAFÍA

Erspamer V., Melchiorri P., Falconieri-Erspamer G., Negri L., Corsi R., Severini C., Barra D., Simmaco M., Kreil G. : a family of naturally occurring peptides with high affinity and selectivity for delta opioid binding sites. Proc Natl Acad Sci U S A. (1989). 86: 5188-5192.

Evans C.J., Keith Jr. D.E., Morrison H., Magendzo K., Edwards R.H., 1992. Cloning of delta opioide receptor by functional expression. Science. (1992). 258: 1952- 1955.

Fang L., Knapp R.J., Horvath R., Matsunaga T.O., Haaseth R.C., Hruby V.J., Porreca F., Yamamura H.I. Characterization of [3H]naltrindole binding to delta opioid receptors in mouse brain and mouse vas deferens: evidence for delta opioid receptor heterogeneity. J Pharmacol Exp Ther. (1994). 268: 836-846.

Faris P.L., Komisaruk B.R., Watkins L.R., Mayer D.J. Evidence for the neuropeptide cholecystokinin as an antagonist of opiate analgesia. Science. (1983). 219: 310- 312.

Faura C.C., Olaso M.J., Garcia Cabanes C., Horga J.F. Lack of morphine-6- glucuronide antinociception after morphine treatment. Is morphine-3- glucuronide involved? Pain. (1996). 65: 25-30.

Fernández-Espejo E. Bases neurobiológicas de la drogadicción. Rev. Neurol. (2002). 34: 659-664.

Flórez J. Fármacos analgésicos opioides. En: Flórez J., Armillo J.A., Mediavilla A. (Eds.). Farmacología humana. 3ª Ed. Masson, Barcelona. (1997): 435-452.

Flórez J., Faura C.C. Farmacología de los analgésicos opioides. En: Aliaga L., Baños J.E., de Barutell C., Molet J., Rodríguez de la Serna A. (Eds.).Tratamiento del dolor teoría y práctica. 2ª Ed. P. Permanyer, Barcelona. (2002): 85-111.

Flórez J., Reig E. Utilización clínica de los opiaceos en el tratamiento del dolor. En: Flórez J., Reig E. (Eds.). Terapéutica farmacológica del dolor. Eunsa, Navarra (1993). 81-117.

215 BIBLIOGRAFÍA

Foley K.M. Changing concepts of tolerante to opioids. En: Chapman C.R., Foley K.M., (Eds). Current and emerging issues in cancer pain: research and practice. Raven Press, New York. (1993). 331-350.

Fowler C.J., Fraser G.L. Mu-, delta-, kappa-opioid receptors and their subtypes. A critical review with emphasis on radioligand binding experiments. Neurochem Int. (1994). 24: 401-426.

France C.P., Medzihradsky F., Woods J.H. Comparison of kappa opioids in rhesus monkeys: behavioral effects and receptor binding affinities. J Pharmacol Exp Ther. (1994). 268: 47-58.

Frances B., Moisand C., Meunier J.C. Na+ ions and Gpp(NH)p selectively inhibit agonist interactions at mu- and kappa-opioid receptor sites in rabbit and guinea- pig cerebellum membranes. Eur J Pharmacol. (1985). 117: 223-232.

Franklin K.B., Abbott F.V. Pentobarbital, diazepam, and ethanol abolish the interphase diminution of pain in the formalin test: evidence for pain modulation by GABAA receptors. Pharmacol Biochem Behav. (1993). 46: 661-666.

Fraser G.L., Labarre M., Godbout C., Butterworth J., Clarke P.B., Payza K., Schmidt R. Characterization of [125I]AR-M100613, a high-affinity radioligand for delta opioid receptors. Peptides. (1999). 20: 1327-1335.

Friedman H.J., Jen M.F., Chang J.K., Lee N.M., Loh H.H. Dynorphin: a possible modulatory peptide on morphine or beta-endorphin analgesia in mouse. Eur J Pharmacol. (1981). 69: 357-360.

Fujimoto J.M., Arts K.S., Rady J.J., Tseng L.F. Spinal (1-17): possible mediator of antianalgesic action. Neuropharmacology. (1990). 29: 609-617.

Funada M., Suzuki T, Narita M., Misawa M., Nagase H. Blockade of morphine reward through the activation of kappa-opioid receptors in mice. Neuropharmacology. (1993). 32: 1315-1323.

216 BIBLIOGRAFÍA

Garner H.R., Burke T.F., Lawhorn C.D., Stoner J.M., Wessinger W.D. - mediated antinociception in mice: partial agonist effects and mu receptor involvement. J Pharmacol Exp Ther. (1997). 282: 1253-1261.

Garzon J., Juarros J.L., Castro M.A., Sanchez-Blazquez P. Antibodies to the cloned mu-opioid receptor detect various molecular weight forms in areas of mouse brain. Mol Pharmacol. (1995). 47: 738-744.

Gelot A., Frances B., Gicquel S., Zajac J.M. Antisense oligonucleotides to human SQA-neuropeptide FF decrease morphine tolerance and dependence in mice. Eur J Pharmacol. (1998). 358: 203-206.

George S.R., Fan T., Xie Z., Tse R., Tam V., Varghese G., O'Dowd B.F. Oligomerization of mu- and delta-opioid receptors. Generation of novel functional properties. J Biol Chem. (2000). 275: 26128-26135.

Gilbert A.K., y Franklin K.B.J. The role of descending fibers from the rostral ventromedial medulla in opioide analgesia in rats. European Journal of Pharmacology. (2002). 449: 75-84.

Gintzler, A.R., Pasternak, G.W. Multiple mu receptors: evidence for mu2 sites in the guinea pig ileum. Neurosci Letters (1983). 39: 51-56.

Girardot M.N., Holloway F.A. Cold water stress analgesia in rats: differential effects of . Physiol Behav. (1994). 32: 547-555.

Goldstein A., Fischli W., Lowney L.I., Hunkapiller M., Hood L. Porcine pituitary dynorphin: complete amino acid sequence of the biologically active heptadecapeptide. Proc Natl Acad Sci U S A. (1981). 78: 7219-7223.

Goldstein A., Tachibana S., Lowney L.I., Hunkapiller M., Hood L. Dynorphin-(1- 13), an extraordinarily potent opioid peptide. Proc Natl Acad Sci U S A. (1979). 76: 6666-6670.

217 BIBLIOGRAFÍA

Gomes I., Gupta A., Filipovska J., Szeto H.H., Pintar J.E., Devi L.A. A role for heterodimerization of mu and delta opiate receptors in enhancing morphine analgesia. Proc Natl Acad Sci U S A. (2004). 101: 5135-5139.

Gomes I., Jordan B.A., Gupta A., Trapaidze N., Nagy V., Devi L.A. Heterodimerization of mu and delta opioid receptors: A role in opiate synergy. J Neurosci. (2000). 20: RC110.

Gong Q.L., Hedner J., Bjorkman R., Hedner T. Morphine-3-glucuronide may functionally antagonize morphine-6-glucuronide induced antinociception and ventilatory depression in the rat. Pain. (1992). 48: 249-255.

Gorman A.L., Elliott K.J., Inturrissi C.E. The d- and l-isomers of methadone bind to the non-competitive site on the N-methil-D-aspartate (NMDA) receptor in rat forebrain and spinal cord. Neurosci. Lett. (1997). 223: 5-8.

Gouarderes C., Faura C.C., Zajac J.M. Rodent strain differences in the NPFF1 and NPFF2 receptor distribution and density in the central nervous system. Brain Res. (2004). 1014: 61-70.

Gouarderes C., Sutak M., Zajac J.M., Jhamandas K. Antinociceptive effects of intrathecally administered F8Famide and FMRFamide in the rat. Eur J Pharmacol. (1993). 237: 73-81.

Gourlay G.K. Different opioids-same actions. En: Kalso E., McQuay H.J., Wiesenfeld-Hallin Z. (Eds.). Opioid sensitivity of chronic noncancer pain. Progress in pain research and management. Vol 14, IASP Press, Seattle, WA. (1999): 97-116.

Grass S., Hoffmann O., Xu X.J., Wiesenfeld-Hallin Z. N-methyl-D-aspartate receptor antagonists potentiate morphine's antinociceptive effect in the rat. Acta Physiol Scand. (1996). 158: 269-273.

218 BIBLIOGRAFÍA

Gray P.A., Rekling J.C., Bocchiaro C.M., Feldman J.L. Modulation of respiratory frequency by peptidergic input to rhythmogenic neurons in the pre-Bötzinger complex. Science. (1999). 286: 1566-1568.

Grevel J., Sadee W. An opiate binding site in the rat brain is highly selective for 4,5- epoxymorphinans. Science. (1983). 221: 1198-1201.

Gross R.A., Moises H.C., Uhler M.D., Macdonald R.L. Dynorphin A and cAMP- dependent protein kinase independently regulate neuronal calcium currents. Proc Natl Acad Sci U S A. (1990). 87: 7025-7029.

Groves P.M., Thompson R.F. Habituation: a dual-process theory. Psychol Rev. (1970). 77: 419-450.

Gucker S., Bidlack J.M. Protein kinase C activation increases the rate and magnitude of agonist-induced delta-opioid receptor down-regulation in NG108-15 cells. Mol Pharmacol. (1992). 42: 656-665.

Guitart X., Beitner-Johnson D., Marby D.W., Kosten T.A., Nestler E.J. Fischer and Lewis rat strains differ in basal levels of neurofilament proteins and their regulation by chronic morphine in the mesolimbic dopamine system. Synapse. (1992). 12: 242-253.

Guitart X., Kogan J.H., Berhow M., Terwilliger R.Z., Aghajanian G.K., Nestler E.J. Lewis and Fischer rat strains display differences in biochemical, electrophysiological and behavioral parameters: studies in the nucleus accumbens and locus coeruleus of drug naive and morphine-treated animals. Brain Res. (1993). 611: 7-17.

Gullapalli S., Ramarao P. L-type Ca2+ channel modulation by dihydropyridines potentiates kappa-opioid receptor agonist induced acute analgesia and inhibits development of tolerance in rats. Neuropharmacology. (2002). 42: 467-475.

219 BIBLIOGRAFÍA

Gutstein H.B., Rubie E.A., Mansour A., Akil H., Woodgett J.R. Opioid effects on mitogen-activated protein kinase signaling cascades. Anesthesiology. (1997). 87: 1118-1126.

Guy E.R., Abbott F.V. The behavioral response to formalin in preweanling rats. Pain. (1992). 51: 81-90.

Hammond D.L. Inference of pain and its modulation from simple behaviours. En: Chapman C.R., Loeser J.D. (Eds.). Issues in pain measurement: advances in pain research and therapy. Vol 12, Raven Press, New York. (1989): 69-91.

Harada H., Ueda H., Katada T., Ui M., Satoh M. Phosphorylated mu-opioid receptor purified from rat brains lacks functional coupling with Gi1, a GTP-binding protein in reconstituted lipid vesicles. Neurosci Lett. (1990). 113: 47-49.

Harrison L.M., Kastin A.J., Zadina J.E. Opiate tolerance and dependence: receptors, G-proteins, and antiopiates. Peptides. (1998). 19: 1603-1630.

Harrison C., Traynor J.R. The [35S]GTPgammaS binding assay: approaches and applications in pharmacology. Life Sci. (2003). 74: 489-508.

Hart S.L., Slusarczyk H., Smith T.W. The involvement of opioid delta-receptors in stress induced antinociception in mice. Eur J Pharmacol. (1983). 95: 283-285.

Hayes R.L., Bennett G.J., Newlon P.G., Mayer D.J. Analgesic effects of certain noxious and stressful manipulations in the rat. Soc. Neurosci. Abstr. (1976) 2: 939.

Hayward M.D., Duman R.S., Nestler E.J. Induction of the c-fos proto-oncogene during opiate withdrawal in the locus coeruleus and other regions of rat brain. Brain Res. (1990). 525: 256-266.

He L., Lee N.M. Delta opioid receptor enhancement of mu opioid receptor-induced antinociception in spinal cord. J Pharmacol Exp Ther. (1998). 285: 1181-1186.

220 BIBLIOGRAFÍA

Hebert T.E., Moffet S., Morello J.P., Loisel T.P., Bichet D.G., Brret C., Bouvier M. A peptide derived from a beta2-adrenergic receptor transmembrane domain inhibits both receptor dimerization and activation. J Biol Chem. (1996). 271: 16384-16392.

Heinricher M.M., Morgan M.M. Mecanismos supraespinales de la analgesia opioide. En: Stein C. (Eds.). Opioides en el control del dolor. Aspectos básicos y clínicos. Masson, Barcelona. (2001): 45-67.

Heinricher M.M., Morgan M.M., Tortorici V, Fields HL. Disinhibition of off-cells and antinociception produced by an opioid action within the rostral ventromedial medulla. Neuroscience. (1994). 63: 279-288.

Heinricher M.M., Tortorici V. Interference with GABA transmission in the rostral ventromedial medulla: disinhibition of off-cells as a central mechanism in nociceptive modulation. Neuroscience. (1994). 63: 533-546.

Henry J.L., Yashpal K., Pitcher G.M., Coderre T.J. Physiological evidence that the 'interphase' in the formalin test is due to active inhibition. Pain. (1999). 82: 57- 63.

Hepburn M.J., Little P.J., Gingras J., Kuhn C.M. Differential effects of naltrindole on morphine-induced tolerance and physical dependence in rats. J Pharmacol Exp Ther. (1997). 281: 1350-1356.

Hepler J.R., Gilman A.G. G proteins. Trends Biochem Sci. (1992). 17: 383-387.

Herman B.H., Goldstein A. Antinociception and paralysis induced by intrathecal dynorphin A. J Pharmacol Exp Ther. (1985). 232: 27-32.

Hernandez M.C., Flores L.R., Bayer B.M. Immunosuppression by morphine is mediated by central pathways. J Pharmacol Exp Ther. (1993). 267: 1336-1341.

Herradon G., Morales L., Alguacil L.F. Differences of mu-opioid receptors between Lewis and F344 rats. Life Sci. (2003). 73: 1537-1542.

221 BIBLIOGRAFÍA

Heyman J.S., Mulvaney S.A., Mosberg H.I., Porreca F. Opioid delta-receptor involvement in supraspinal and spinal antinociception in mice. Brain Res. (1987). 420: 100-108.

Heyman J.S., Vaught J.L., Mosberg H.I., Haaseth R.C., Porreca F. Modulation of mu-mediated antinociception by delta agonists in the mouse: selective

potentiation of morphine and by [D-Pen2,D-Pen5]enkephalin. Eur J Pharmacol. (1989). 165: 1-10.

Hoffmann O., Plesan A., Wiesenfeld-Hallin Z. Genetic differences in morphine sensitivity, tolerance and withdrawal in rats. Brain Res. (1998). 806: 232-237.

Hoffmann O., Wiesenfeld-Hallin Z. Dextromethorphan potentiates morphine antinociception, but does not reverse tolerance in rats. Neuroreport. (1996). 7: 838-840.

Hole K., Tjolsen A. The tail-flick and formalin tests in rodents: changes in skin temperature as a confounding factor. Pain. (1993). 53: 247-254.

Hong E.J., Rice K.C., Calderon S., Woods J.H., Traynor J.R. Convulsive behavior of nonpeptide delta-opioid ligands: comparison of SNC80 and BW373U86 in mice. Analgesia. (1998). 3: 269-276.

Horan P., Tallarida R.J., Haaseth RC, Matsunaga TO, Hruby VJ, Porreca F. Antinociceptive interactions of opioid delta receptor agonists with morphine in mice: supra- and sub-additivity. Life Sci. (1992). 50: 1535-1541.

Horvath G. -1 and endomorphin-2: pharmacology of the selective endogenous mu-opioid receptor agonists. Pharmacol Ther. (2000). 88: 437-463.

Hosohata Y., Vanderah T.W., Burkey T.H., Ossipov M.H., Kovelowski C.J., Sora I., Uhl G.R., Zhang X., Rice K.C., Roeske W.R., Hruby V.J., Yamamura H.I., Lai J., Porreca F. Delta-Opioid receptor agonists produce antinociception and [35S]GTPgammaS binding in mu receptor knockout mice. Eur J Pharmacol. (2000). 388: 241-248.

222 BIBLIOGRAFÍA

Hsia J.A., Moss J., Hewlett E.L., Vaughan M. ADP-ribosylation of adenylate cyclase by pertussis toxin. Effects on inhibitory agonist binding. J Biol Chem. (1984). 259: 1086-1090.

Hughes J., Smith T.W., Kosterlitz H.W., Fothergill L.A., Morgan B.A., Morris H.R. Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature. (1975). 258: 577-580.

Hunskaar S., Hole K. The formalin test in mice: dissociation between inflammatory and non-inflammatory pain. Pain. (1987). 30: 103-114.

Hurley R.W., Hammond D.L. Contribution of endogenous to the enhanced analgesic effects of supraspinal mu opioid receptor agonists after inflammatory injury. J Neurosci. (2001). 21: 2536-2545.

Ikeda K., Ichikawa T., Kobayashi T., Kumanishi T., Oike S., Yano R. Unique behavioural phenotypes of recombinant-inbred CXBK mice: partial deficiency of sensitivity to mu- and kappa-agonists. Neurosci Res. (1999). 34: 149-155.

Ingram S.L., Williams J.T. Opioid inhibition of Ih via adenylyl cyclase. Neuron. (1994). 13: 179-186.

Ingram S.L., Williams J.T. Modulation of the hyperpolarization-activated current (Ih) by cyclic nucleotides in guinea-pig primary afferent neurons. J Physiol. (1996). 492: 97-106.

Irwin S., Houde R.W., Bennett D.R., Hendershot L.C., Seevers M.H. The effects of morphine methadone and meperidine on some reflex responses of spinal animals to nociceptive stimulation. J Pharmacol Exp Ther. (1951). 101: 132- 143.

Izenwasser S., Buzas B., Cox B.M. Differential regulation of adenylyl cyclase activity by mu and delta opioids in rat caudate putamen and nucleus accumbens. J Pharmacol Exp Ther. (1993). 267: 145-152.

223 BIBLIOGRAFÍA

Izenwasser S., Staley J.K., Cohn S., Mash D.C. Characterization of kappa1-opioid receptor binding in human insular cortex. Life Sci. (1999). 65: 857-862.

Jackson H.C., Kitchen I. Swim-stress induced antinociception in young rats. Br. J. Pharmacol. (1989). 96: 617-622.

Jaffe J.H., Martin WR. Opioid and antagonist. En: Hardman JG, Limbird LE, Molinoff PB, Ruddon RW y Goodman GA (Eds.). The pharmacological basis of therapeutics. Goodman and Gilman´s (1992). 479-512.

Jagerovic N., Cano C., Elguero J., Goya P., Callado L.F., Meana J.J., Giron R., Abalo R., Ruiz D., Goicoechea C., Martin M.A. Long-acting fentanyl analogues: synthesis and pharmacology of N-(1-phenylpyrazolyl)-N-(1- phenylalkyl-4-piperidyl)propanamides. Bioorg Med Chem. (2002). 10: 817- 827.

Jang Y., Yoburn B.C. Evaluation of receptor mechanism mediating fentanyl analgesia and toxicity. Eur J Pharmacol. (1991). 197: 135-141.

Jiang Q.J., Mosberg H.I., Porreca F. Modulation of the potency and efficacy of mu- mediated antinociception by delta agonists in the mouse. J Pharmacol Exp Ther. (1990). 254: 683-689.

Jiang Q., Takemori A.E., Sultana M., Portoghese P.S., Bowen W.D., Mosberg H.I., Porreca F. Differential antagonism of opioide delta antinociception by [D-

Ala2,Leu5,Cys6*enkephalin and naltrindole 5´-isothiocyanate: evidence for delta receptor subtypes. J Pharmacol Exp Ther. (1991). 257: 1069-1075.

Johnson S.W., North R.A. Opioids excite dopamine neurons by hyperpolarization of local interneurons. J Neurosci. (1992). 12: 483-488.

Jordan B.A., Devi L.A. G-protein-coupled receptor heterodimerization modulates receptor function. Nature. (1999). 399: 697-700.

224 BIBLIOGRAFÍA

Jourdan D., Ardid D., Bardin L., Bardin M., Neuzeret D., Lanphouthacoul L., Escalier A. A new automated method of pain scoring in the formalin test in rats. Pain. (1997). 71: 265-270.

Jutkiewicz E.M., Eller E.B., Folk J.E., Rice K.C., Traynor J.R., Woods J.H. Delta- opioid agonists: differential efficacy and potency of SNC80, its 3-OH (SNC86) and 3-desoxy (SNC162) derivatives in Sprague-Dawley rats. J Pharmacol Exp Ther. (2004). 309: 173-181.

Kakidani H., Furutani Y., Takahashi H., Noda M., Morimoto Y., Hirose T., Asai M., Inayama S., Nakanishi S., Numa S. Cloning and sequence analysis of cDNA for porcine beta-neo-endorphin/dynorphin precursor. Nature. (1982). 298: 245- 249.

Kanarek R.B., Przypek J., D'Anci K.E., Marks-Kaufman R. Dietary modulation of mu and kappa opioid receptor-mediated analgesia. Pharmacol Biochem Behav. (1997). 58: 43-49.

Kellstein D.E., Mayer D.J. Spinal co-administration of cholecystokinin antagonists with morphine prevents the development of opioid tolerance. Pain. (1991). 47: 221-229.

Kest B., Beczkowska I., Franklin S.O., Lee C.E., Mogil J.S., Inturrisi C.E. Differences in delta opioid receptor antinociception, binding, and mRNA levels between BALB/c and CXBK mice. Brain Res. (1998). 805: 131-137.

Kest B., Hopkins E., Palmese C.A., Adler M., Mogil J.S. Genetic variation in morphine analgesic tolerance: A survey of 11 inbred mouse strains. Pharmacol. Biochem. Behav. (2002). 73: 821-828.

Kest B., Jenab S., Brodsky M., Sadowski B., Belknap J.K., Mogil J.S., Inturrisi C.E. Mu and delta opioid receptor analgesia, binding density, and mRNA levels in mice selectively bred for high and low analgesia. Brain Res. (1999). 816: 381- 389.

225 BIBLIOGRAFÍA

Kest B., Lee C.E., McLemore G.L., Inturrisi C.E. An antisense oligodeoxynucleotide to the delta opioid receptor (DOR-1) inhibits morphine tolerance and acute dependence in mice. Brain Res Bull. (1996). 39: 185-188.

Kest B., McLemore G.L., Sadowski B., Mogil J.S., Belknap J.K., Inturrisi C.E. Acute morphine dependence in mice selectively-bred for high and low analgesia. Neurosci Lett. (1998b). 256:120-122.

Kest B., Mogil J.S., Sternberg W.F., Liebeskind J.C., Sadowski B. Evidence for the up-regulation of kappa opiate mechanisms in mice selectively bred for high analgesia. Proc West Pharmacol Soc. (1993). 36: 249-253.

Kest B., Palmese C.A., Hopkins E., Adler M., Juni A., Mogil J.S. Naloxone- precipitated withdrawal jumping in 11 inbred mouse strains: evidence for common genetic mechanisms in acute and chronic morphine physical dependence. Neuroscience. (2002b). 115: 463-469.

Kiefel J.M., Rossi G.C., Bodnar R.J. Medullary mu and delta opioide receptors modulate mesencephalic morphine analgesia in rats. Brain Res. (1993). 624: 151-161.

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

Kieffer B.L., Befort K., Gaveriaux-Ruff C., Hirth C.G. The delta-opioid receptor: isolation of a cDNA by expression cloning and pharmacological characterization. Proc Natl Acad Sci U S A. (1992). 89: 12048-12052.

Kieffer B.L., Gaveriaux-Ruff C. Exploring the opioid system by gene knockout. Prog Neurobiol. (2002). 66: 285-306.

Kitchen I., Pinker S.R. Antagonism of swim-stress-induced antinociception by the delta-opioid receptor antagonist naltrindole in adult and young rats. Br J Pharmacol. (1990). 100: 685-688.

226 BIBLIOGRAFÍA

Knapp R.J., Sharma S.D., Toth G., Duong M.T., Fang L., Bogert C.L., Weber S.J.,

125 Hunt M., Davis T.P., Wamsley J.K., Hruby J.V., Yamamura H.I. [D-Pen2,4'- I-

Phe4,D-Pen5]Enkephalin: a selective high affinity radioligand for delta opioid receptors with exceptional specific activity. J Pharmacol Exp Ther. (1991). 258: 1077-1083.

Ko M.C., Lee H., Harrison C., Clark M.J., Song H.F., Naughton N.N., Woods J.H., Traynor J.R. Studies of mu-, kappa-, and delta-opioid receptor density and G protein activation in the cortex and thalamus of monkeys. J Pharmacol Exp Ther. (2003). 306:179-186.

Kohno M., Fukushima N., Yoshida A., Ueda H. G(i1) and G(oA) differentially determine kinetic efficacies of agonists for kappa-opioid receptor. FEBS Lett. (2000). 473: 101-105.

König M., Zimmer A.M., Steiner H., Holmes P.V., Crawley J.N., Brownstein M.J., Zimmer A. Pain responses, anxiety and aggression in mice deficient in pre- . Nature. (1996). 383: 535-538.

Koob G.F., Le Moal M. Drug abuse: hedonic homeostatic dysregulation. Science. (1997). 278: 52-58.

Koob G.F., Maldonado R., Stinus L. Neural substrates of opiate withdrawal. Trends Neurosci. (1992). 15: 186-191.

Kopin A.S., McBride E.W., Gordon M.C., Quinn S.M., Beinborn M. Inter- and intraspecies polymorphisms in the cholecystokinin-B/gastrin receptor alter drug efficacy. Proc Natl Acad Sci U S A. (1997). 94:11043-11048.

Kosten T.A., Ambrosio E. HPA axis function and drug addictive behaviors: insights from studies with Lewis and Fischer 344 inbred rats. Psychoneuroendocrinology. (2002). 27: 35-69.

227 BIBLIOGRAFÍA

Kosterlitz H.W., Corbett A.D., Gillan M.G., McKnight A.T., Paterson S.J., Robson L.E. Recent developments in bioassay using selective ligands and selective in vitro preparations. NIDA Res Monogr. (1986). 70: 223-235.

Kovelowski C.J., Bian D., Hruby V.J., Lai J., Ossipov M.H., Porreca F. Selective opioid delta agonists elicit antinociceptive supraspinal/spinal synergy in the rat. Brain Res. (1999). 843: 12-17.

LaBuda C.J., Sora I., Uhl G.R., Fuchs P.N. Stress-induced analgesia in mu-opioid receptor knockout mice reveals normal function of the delta-opioid receptor system. Brain Res. (2000). 869: 1-5.

Lahti R.A., Mickelson M.M., McCall J.M., Von Voigtlander P.F. [3H]U-69593 a highly selective ligand for the opioid kappa receptor. Eur J Pharmacol. (1985). 109: 281-284.

Lake J.R., Hebert K.M., Payza K., Deshotel K.D., Hausam D.D., Witherspoon W.E., Arcangeli K.A., Malin D.H. Analog of neuropeptide FF attenuates morphine tolerance. Neurosci Lett. (1992). 146: 203-206.

Lake J.R., Hammond M.V., Shaddox R.C., Hunsicker L.M., Yang H.Y., Malin D.H. IgG from neuropeptide FF antiserum reverses morphine tolerance in the rat. Neurosci Lett. (1991). 132: 29-32.

Lalley P.M. Mu-opioid receptor agonist effects on medullary respiratory neurons in the cat: evidence for involvement in certain types of ventilatory disturbances. Am J Physiol Regul Integr Comp Physiol. (2003). 285: R1287-1304.

Lane-Ladd S.B., Pineda J., Boundy V.A., Pfeuffer T., Krupinski J., Aghajanian G.K., Nestler E.J. CREB (cAMP response element-binding protein) in the locus coeruleus: biochemical, physiological, and behavioral evidence for a role in opiate dependence. J Neurosci. (1997). 17: 7890-7901.

228 BIBLIOGRAFÍA

Langerman L., Zakowski M.I., Piskoun B., Grant G.J. Hot plate versus tail flick: evaluation of acute tolerance to continuous morphine infusion in the rat model. J Pharmacol Toxicol Methods. (1995). 34: 23-27.

Larson A.A., Vaught J.L., Takemori A.E. The potentiation of spinal analgesia by leucine enkephalin. Eur J Pharmacol. (1980). 61: 381-383.

Lasagna L., Beecher H.K. The optimal dose of morphine. J Am Med Assoc. (1954). 156: 230-234.

Laulin J.P., Celerier E., Larcher A., Le Moal M., Simonnet G. Opiate tolerance to daily heroin administration: an apparent phenomenon associated with enhanced pain sensitivity. Neuroscience. (1999). 89: 631-636.

Law P.Y., Hom D.S., Loh H.H. Loss of opiate receptor activity in neuroblastoma X glioma NG108-15 hybrid cells after chronic opiate treatment. A multiple-step process. Mol Pharmacol. (1982). 22: 1-4.

Law P.Y., Wong Y.H., Loh H.H. Molecular mechanisms and regulation of opioid receptor signaling. Annu Rev Pharmacol Toxicol. (2000). 40: 389-430.

Lazarus L.H., Bryant S.D., Attila M., Salvadori S. Frog skin opioid peptides: a case for environmental mimicry. Environ Health Perspect. (1994). 102: 648-654.

Le Bars D., Gozariu M., Cadden S.W. Animal models of nociception. Pharmacol Rev. (2001). 53: 597-652.

Lee N.M., Leybin L., Chang J.K., Loh H.H. Opiate and peptide interaction: effect of enkephalins on morphine analgesia. Eur J Pharmacol. (1980). 68: 181-185.

Lefkowitz R.J., Stadel J.M., Caron M.G. Adenylate cyclase-coupled beta-adrenergic receptors: structure and mechanisms of activation and desensitization. Annu Rev Biochem. (1983). 52: 159-186.

229 BIBLIOGRAFÍA

Leone P., Pocock D., Wise R.A. Morphine-dopamine interaction: ventral tegmental morphine increases nucleus accumbens dopamine release. Pharmacol Biochem Behav. (1991). 39: 469-472.

Levac B.A., O'Dowd B.F., George S.R. Oligomerization of opioid receptors: generation of novel signaling units. Curr Opin Pharmacol. (2002). 2: 76-81.

Levine J.D., Gordon N.C. Synergism between the analgesic actions of morphine and pentazocine. Pain. (1988). 33: 369-372.

Lewis, J.D., Khachaturian, H., Watson, S.J., Comparative distribution of opiate receptors and three opioide peptide neuronal systems in rhesus monkey central nervous system. Life Sciences (1983). 33: 239-242.

Li C.H., Chung D. Isolation and structure of an untriakontapeptide with opiate activity from camel pituitary glands. Proc Natl Acad Sci U S A. (1976). 73: 1145-1148.

Liu J.G., Anand K.J. Protein kinases modulate the cellular adaptations associated with opioid tolerance and dependence. Brain Res Brain Res Rev. (2001). 38: 1- 19.

Lord J.A.H., Waterfield A.A., Hughes J., Kosterlitz H.W. Endogenous opioide peptides: multiple agonists and receptors. Nature. (1977). 267: 495-499.

Lou L.G., Pei G. Modulation of protein kinase C and cAMP-dependent protein kinase by delta-opioid. Biochem Biophys Res Commun. (1997). 236: 626-629.

Lu L., Huang M., Liu Z., Ma L. Cholecystokinin-B receptor antagonists attenuate morphine dependence and withdrawal in rats. Neuroreport. (2000). 11: 829- 832.

Lutfy K., Chang S.C., Candido J., Jang Y, Sierra V., Yoburn B.C. Modification of morphine-induced analgesia and toxicity by pertussis toxin. Brain Res. (1991). 544: 191-195.

230 BIBLIOGRAFÍA

Lutfy K., Sadowski B., Kwon I.S., Weber E. Morphine analgesia and tolerance in mice selectively bred for divergent swim stress-induced analgesia. Eur J Pharmacol. (1994). 265: 171-174.

Lutfy K., Sadowski B., Marek P., Kwon I.S., Keana J.F., Weber E. Differential sensitivity of mice bred for stress-induced analgesia to morphine and ACEA- 1011 in the formalin test. Pharmacol Biochem Behav. (1996). 54: 495-500.

Lysle D.T., Coussons M.E., Watts V.J., Bennett E.H., Dykstra L.A. Drug of abuse, inmunity and AIDS. En: Friedman H., Klein T., Spector S. (Eds.). Advances in Experimental Medicine and Biology. Plenum Press, New York (1993). 335: 53- 59.

Maeda T., Kishioka S., Inoue N., Shimizu N., Fukazawa Y., Ozaki M., Yamamoto H. Naloxone-precipitated morphine withdrawal elicits increases in c-fos mRNA expression in restricted regions of the infant rat brain. Jpn J Pharmacol. (2002). 90: 270-275.

Maher C.E., Selley D.E., Childers S.R. Relationship of mu opioid receptor binding to activation of G-proteins in specific rat brain regions. Biochem Pharmacol. (2000). 59: 1395-1401.

Malin D.H., Lake J.R., Fowler D.E., Hammond M.V., Brown S.L., Leyva J.E., Prasco P.E., Dougherty T.M. FMRF-NH2-like mammalian peptide precipitates opiate-withdrawal syndrome in the rat. Peptides. (1990). 11: 277-280.

Malin D.H., Lake J.R., Hammond M.V., Fowler D.E., Rogillio R.B., Brown S.L., Sims J.L., Leecraft B.M., Yang H.Y. FMRF-NH2-like mammalian octapeptide: possible role in opiate dependence and abstinence. Peptides. (1990b). 11: 969- 972.

Malmberg A.B., Yaksh T.L. Isobolographic and dose-response analyses of the interaction between intrathecal mu and delta agonists: effects of naltrindole and its benzofuran analog (NTB). J Pharmacol Exp Ther. (1992). 263: 264-275.

231 BIBLIOGRAFÍA

Maldonado R., Negus S., Koob G.F. Precipitation of morphine withdrawal syndrome in rats by administration of mu-, delta- and kappa-selective opioid antagonists. Neuropharmacology. (1992). 31: 1231-1241.

Maldonado R., Valverde O. Participation of the opioid system in cannabinoid- induced antinociception and emotional-like responses. Eur Neuropsychopharmacol. (2003). 13: 401-410.

Maldonado R., Valverde O., Ducos B., Blommaert A.G., Fournie-Zaluski M.C., Roques B.P. Inhibition of morphine withdrawal by the association of RB 101, an inhibitor of enkephalin catabolism, and the CCKB antagonist PD-134,308. Br J Pharmacol. (1995). 114: 1031-1039.

Malin D.H., Lake J.R., Arcangeli K.R., Deshotel K.D., Hausam D.D., Witherspoon W.E., Carter V.A., Yang H.Y., Pal B., Burgess K. Subcutaneous injection of an analog of neuropeptide FF precipitates morphine abstinence syndrome. Life Sci. (1993). 53: PL261-266.

Manzanares J., Corchero J., Romero J., Fernandez-Ruiz J.J., Ramos J.A., Fuentes J.A. Pharmacological and biochemical interactions between opioids and cannabinoids. Trends Pharmacol Sci. (1999). 20: 287-294.

Marek P., Ben-Eliyahu S., Gold M., Liebeskind J.C. Excitatory amino acid antagonists (kynurenic acid and MK-801) attenuate the development of morphine tolerance in the rat. Brain Res. (1991). 547: 77-81.

Marek P., Mogil J.S., Sternberg W.F., Panocka I., Liebeskind J.C. N-methyl-D- aspartic acid (NMDA) receptor antagonist MK-801 blocks non-opioid stress- induced analgesia. II. Comparison across three swim-stress paradigms in selectively bred mice. Brain Res. (1992). 578: 197-203.

Marley R.J., Elmer G.I. and Goldberg S.R. The use of pharmacogenetic techniques in drug abuse research. Pharmacol Ther. (1992). 53: 217– 237.

232 BIBLIOGRAFÍA

Marki A., Monory K., Otvos F., Toth G., Krassnig R., Schmidhammer H., Traynor J.R., Roques B.P., Maldonado R., Borsodi A. Mu-opioid receptor specific antagonist : characterization by in vitro radioligand and [35S]GTPgammaS binding assays. Eur J Pharmacol. (1999). 383: 209-214.

Martin W.R., Eades C.G., Thompson J.A., Huppler R.E., Gilbert P.E. The effects of morphine and -like drugs in the nondependent and morphine- dependent chronic spinal dog. J Pharmal Exp Ther. (1976). 197: 517-532.

Martin S., Lyupina Y., Crespo J.A., Gonzalez B., Garcia-Lecumberri C., Ambrosio E. Genetic differences in NMDA and D1 receptor levels, and operant responding for food and morphine in Lewis and Fischer 344 rats. Brain Res. (2003). 973: 205-213.

Martin S., Manzanares J., Corchero J., Garcia-Lecumberri C., Crespo J.A., Fuentes J.A., Ambrosio E. Differential basal proenkephalin gene expression in dorsal striatum and nucleus accumbens, and vulnerability to morphine self- administration in Fischer 344 and Lewis rats. Brain Res. (1999). 821: 350-355.

Martin M., Matifas A., Maldonado R., Kieffer B.L. Acute antinociceptive responses in single and combinatorial opioid receptor knockout mice: distinct mu, delta and kappa tones. Eur J Neurosci. (2003). 17: 701-708.

Martin N.A., Prather P.L. Interaction of co-expressed mu- and delta-opioid receptors in transfected rat pituitary GH(3) cells. Mol Pharmacol. (2001). 59: 774-783.

Mas M., Sabater E., Olaso M.J., Horga J.F., Faura C.C. Genetic variability in morphine sensitivity and tolerance between different strains of rats. Brain Res. (2000). 866: 109-115.

Mather L.E., Smith M.T. Farmacología clínica y efectos adversos. En: Stein C. (Eds.). Opioides en el control del dolor. Aspectos básicos y clínicos. Masson, Barcelona. (2001): 183-205.

233 BIBLIOGRAFÍA

Matsuoka I., Maldonado R., Defer N., Noel F., Hanoune J., Roques B.P. Chronic morphine administration causes region-specific increase of brain type VIII adenylyl cyclase mRNA. Eur J Pharmacol. (1994). 268: 215-221

Matthes H.W., Maldonado R., Simonin F., Valverde O., Slowe S., Kitchen I., Befort K., Dierich A., Le Meur M., Dolle P., Tzavara E., Hanoune J., Roques BP., Kieffer BL. Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene. Nature. (1996). 383: 819-823.

Matthes H.W., Smadja C., Valverde O., Vonesch J.L., Foutz A.S., Boudinot E., Denavit-Saubie M., Severini C., Negri L., Roques B.P., Maldonado R., Kieffer B.L. Activity of the delta-opioid receptor is partially reduced, whereas activity of the kappa-receptor is maintained in mice lacking the mu-receptor. J Neurosci. (1998). 18: 7285-7295.

Mattia A., Vanderah T., Mosberg H.I., Porreca F. Lack of antinociceptive cross-

tolerance betweem [D-Pen2, D-Pen5]enkephalin and [D-Ala2]deltorphin II in mice: evidence for delta receptor subtypes. J Pharmacol Exp Ther. (1991). 258: 583-587.

Mattia, A., Farmer, S.C., Takemori, A.E., Sultana, M., Portoghese, P.S., Mosberg., H.I., Bowen, W.D., Porreca, F. Spinal opioide delta antinociception in the mouse: mediation by a 5´-NTII-sensitive delta receptor subtype. J Pharmacol Expl Ther. (1992). 260: 518-525.

Mauborgne A., Bourgoin S., Polienor H., Roumy M., Simonnet G., Zajac J.M., Cesselin F. The neuropeptide FF analogue, 1DMe, acts as a functional opioid autoreceptor antagonist in the rat spinal cord. Eur J Pharmacol. (2001). 430: 273-276.

Mayer D.J., Price D.D. Central nervous system mechanisms of analgesia. Pain. (1976). 2: 379-404.

234 BIBLIOGRAFÍA

Mayer P., Schulzeck S., Kraus J., Zimprich A., Hollt V. Promoter region and alternatively spliced exons of the rat mu-opioid receptoe gene. Journal of Neurochemistry. (1996). 66: 2272-2278.

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

McCleane G.J. The cholecystokinin antagonist enhances the analgesic efficacy of morphine in humans with chronic benign pain. Anesth Analg. (1998). 87: 1117-1120.

McCrimmon D.R., Alheid G.F. On the opiate trail of respiratory depression. Am J Physiol Regul Integr Comp Physiol. (2003). 285:R1274-1275.

McKnight A.T., Corbett A.D., Marcoli M., Kosterlitz H.W. The opioid receptors in the hamster vas deferens are of the delta-type. Neuropharmacology. (1985). 24: 1011-1017.

McLaughlin J.P., Marton-Popovici M., Chavkin C. Kappa opioid receptor antagonism and gene disruption block stress-induced behavioral responses. J Neurosci. (2003). 23: 5674-5683.

McQuay H.J. Pharmacological treatment of neuralgic and neuropathic pain. Cancer Surv. (1988). 7: 141-159.

McQuay H.J. Opioid in pain management. Lancet (2001). 357: 37-45.

Mehmanesh H., Almeida O.F., Nikolarakis K.E., Herz A. Hypothalamic LH-RH release after acute and chronic treatment with morphine studied in a combined in vivo/in vitro model. Brain Res. (1988). 451: 69-76.

Menendez L., Andres-Trelles F., Hidalgo A., Baamonde A. Involvement of spinal kappa opioid receptors in a type of footshock induced analgesia in mice. Brain Res. (1993). 611: 264-271.

235 BIBLIOGRAFÍA

Meng F., Xie G.X., Thompson R.C., Mansour A., Goldstein A., Watson S.J., Akil H. Cloning and pharmacological characterization of a rat kappa opioid receptor. Proc Natl Acad Sci U S A. (1993). 90: 9954-9958.

Miaskowski C., Sutters K.A., Taiwo Y.O., Levine J.D. Antinociceptive and motor effects of delta/mu and kappa/mu combinations of intrathecal opioid agonists. Pain. (1992). 49: 137-144.

Miaskowski C., Taiwo Y.O., Levine J.D. Kappa- and delta-opioid agonists synergize to produce potent analgesia. Brain Res. (1990). 509: 165-168.

Millan M.J., Czlonkowski A., Lipkowski A., Herz A. Kappa-opioid receptor- mediated antinociception in the rat. II. Supraspinal in addition to spinal sites of action. J Pharmacol Exp Ther. (1989). 251: 342-350.

Minamino N., Kangawa K., Chino N., Sakakibara S., Matsuo H. Beta-neo- endorphin, a new hypothalamic "big" Leu-enkephalin of porcine origin: its purification and the complete amino acid sequence. Biochem Biophys Res Commun. (1981). 99: 864-870.

Mitchell J.M., Basbaum A.I., Fields H.L. A locus and mechanism of action for associative morphine tolerance. Nat Neurosci. (2000). 3: 47-53.

Mizoguchi H., Leitermann R.J., Narita M., Nagase H., Suzuki T., Tseng L.F. Region- dependent G-protein activation by kappa-opioid receptor agonists in the mouse brain. Neurosci Lett. (2004). 356: 145-147.

Mizoguchi H., Narita M., Kampine J.P., Tseng L.F. [Met5]enkephalin and delta2- opioid receptors in the spinal cord are involved in cold water swimming- induced antinociception in the mouse. Life Sci. (1997). 61: PL81-6.

Mizoguchi H., Wu H.E., Narita M., Sora I., Hall S.F., Uhl G.R., Loh H.H., Nagase H., Tseng L.F. 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. (2003). 93: 423-429.

236 BIBLIOGRAFÍA

Mogil J.S. The genetic mediation of individual differences in sensitivity to pain and its inhibition. Proc Natl Acad Sci U S A. (1999). 96: 7744-7751.

Mogil J.S., Belknap J.K. Sex and genotype determine the selective activation of neurochemically-distinct mechanisms of swim stress-induced analgesia. Pharmacol Biochem Behav. (1997). 56: 61-66.

Mogil J.S., Grisel J.E., Hayward M.D., Bales J.R., Rubinstein M., Belknap J.K., Low M.J. Disparate spinal and supraspinal opioid antinociceptive responses in beta- endorphin-deficient mutant mice. Neuroscience. (2000). 101: 709-717.

Mogil J.S., Marek P., Flodman P., Spence M.A., Sternberg W.F., Kest B., Sadowski B., Liebeskind J.C. One or two genetic loci mediate high opiate analgesia in selectively bred mice. Pain. (1995). 60: 125-135.

Mogil J.S., Marek P., O'Toole L.A., Helms M.L., Sadowski B., Liebeskind J.C., Belknap J.K. Mu-opiate receptor binding is up-regulated in mice selectively bred for high stress-induced analgesia. Brain Res. (1994). 653: 16-22.

Mogil J.S., Pasternak G.W. The molecular and behavioural pharmacology of the orphanin FQ/ peptide and receptor family. Pharmacol Rev. (2001). 53: 381-415.

Mogil J.S., Richards S.P., O'Toole L.A., Helms M.L., Mitchell S.R., Belknap J.K. Genetic sensitivity to hot-plate nociception in DBA/2J and C57BL/6J inbred mouse strains: possible sex-specific mediation by delta2-opioid receptors. Pain. (1997). 70: 267-277.

Mogil J.S., Sternberg W.F., Marek P., Sadowski B., Belknap J.K., Liebeskind J.C. The genetics of pain and pain inhibition. Proc Natl Acad Sci U S A. (1996). 93: 3048-3055.

Mogil J.S., Wilson S.G., Bon K., Lee S.E., Chung K., Raber P., Pieper J.O., Hain H.S., Belknap J.K., Hubert L., Elmer G.I., Cheng J.M., Devor M. Heritability

237 BIBLIOGRAFÍA

of nociception II. “Types” of nociception revealed by genetic correlation análisis. Pain (1999). 80: 83-93.

Morgan D., Cook C.D., Picker M.J. Sensitivity to the discriminative stimulus and antinociceptive effects of mu opioids: role of strain of rat, stimulus intensity, and intrinsic efficacy at the mu opioid receptor. J Pharmacol Exp Ther. (1999). 289: 965-975.

Morin-Surun M.P., Boudinot E., Gacel G., Champagnat J., Roques B.P., Denavit- Saubie M. Different effects of mu and delta opiate agonists on respiration. Eur J Pharmacol. (1984). 98: 235-240.

Morre M., Bachy A., Gout B., Boigegrain R., Arnone M., Roncucci R. Kappa binding sites in guinea-pig brain membranes: evidence for a dynorphin- resistant subtype. Life Sci. (1983). 33 (Suppl 1): 179-182.

Moskowitz A.S., Goodman R.R. Autoradiographic distribution of mu1 and mu2 opioid binding in the mouse central nervous system. Brain Research (1985). 360: 117-129.

Moskowitz A.S., Terman G.W., Liebeskind J.C. Stress-induced analgesia in the mouse: strain comparisons. Pain. (1985b). 23: 67-72.

Motulsky H.J. Two-way analysis of variance, In: Analyzing Data with GraphPad Prism. GraphPad Software Inc., San Diego CA, www.graphpad.com. (1999): 89-103.

Muhammad B.Y., Kitchen I. Effect of delayed weaning on opioid receptor control of swim stress-induced antinociception in the developing rat. Br J Pharmacol. (1993). 109: 651-654.

Nagase C.S., Randich A., Brush F.R. Genetic differences in avoidance behavior: cardiovascular activity, pain sensitivity and stress-induced analgesia. Peptides. (1985). 6 (Suppl 1): 29-35.

238 BIBLIOGRAFÍA

Nakanishi S., Inoue A., Kita T., Nakamura M., Chang A.C., Cohen S.N., Numa S. Nucleotide sequence of cloned cDNA for bovine corticotropin-beta- precursor. Nature. (1979). 278: 423-427.

Narita M., Funada M., Suzuki T. Regulations of opioid dependence by opioid receptor types. Pharmacol Ther. (2001). 89: 1-15.

Narita M., Imai S., Ozaki S., Suzuki M., Narita M., Suzuki T. Reduced expression of a novel mu-opioid receptor (MOR) subtype MOR-1B in CXBK mice: implications of MOR-1B in the expression of MOR-mediated responses. Eur J Neurosci. (2003). 18: 3193-3198.

Nebert D.W., Weber W.W. Pharmacogenetics, En: Pratt W.B., Taylor (Eds.). Principles of drug action. The basis of Pharmacology. Churchill Livingstone, New York. (1990):.469-531.

Neilan C.L., King M.A., Rossi G., Ansonoff M., Pintar J.E., Schiller P.W., Pasternak G.W. Differential sensitivities of mouse strains to morphine and [Dmt1]DALDA analgesia. Brain Res. (2003). 974: 254-257.

Ness T.J. y Gebhard G.F. Centrifugal modulation of the rat tail flick reflex evoked by graded noxious heating of the tail, Brain Res. (1986). 386: 41-52.

Nestler E.J., Aljera M., Aghajanian G.K. Molecular Control of Locus Coeruleus Neurotransmission. Biological Psychiatry. (1999). 46: 1131-1139.

Ng, G.Y., O´Dowd, B.F., Lee, S.P., Chung, H.T., Brann, M.R., Seeman, P., George, S.R. Dopamine D2 receptor dimers and receptor-blocking peptides. Biochemical and Biophysical Research Communications. (1996). 277: 200- 204.

Nichols M.L., Bian D., Ossipov M.H., Malan T.P. Jr., Porreca F. Antiallodynic effects of a CCKB antagonist in rats with nerve ligation injury: role of endogenous enkephalins. Neurosci Lett. (1996). 215: 161-164.

239 BIBLIOGRAFÍA

Nijssen P.C., Sexton T., Childers S.R. Opioid-inhibited adenylyl cyclase in rat brain membranes: lack of correlation with high-affinity opioid receptor binding sites. J Neurochem. (1992). 59: 2251-2262.

Nimchinsky E.A., Hof P.R., Janssen W.G. Morrison J.H., Schmauss C. Expresión of dopamine D3 receptors dimers and tetramers in brain and in transfected cells. Journal of Biological Chemistri. (1997). 272: 29229-29237.

Nitsche J.F., Schuller A.G.P., King M.A., Zengh M., Pasternak G.W., Pintar J.E. Genetic dissociation of opiate tolerance and physical dependence in δ-opioid receptor-1 and prepro enkephalin knock-out mice. J. Neurosci. (2002). 22: 10906-10913.

Nock B., Giordano A.L., Cicero T.J., O'Connor L.H. Affinity of drugs and peptides for U-69,593-sensitive and -insensitive kappa opiate binding sites: the U- 69,593-insensitive site appears to be the beta endorphin-specific epsilon receptor. J Pharmacol Exp Ther. (1990). 254: 412-419.

Nock B., Giordano A.L., Moore B.W., Cicero T.J. Properties of the putative epsilon opioid receptor: identification in rat, guinea pig, cow, pig and chicken brain. J Pharmacol Exp Ther. (1993). 264: 349-359.

Noda M., Furutani Y., Takahashi H., Toyosato M., Hirose T., Inayama S., Nakanishi S., Numa S. Cloning and sequence analysis of cDNA for bovine adrenal preproenkephalin. Nature. (1982). 295: 202-206.

North R.A., Williams J.T., Surprenant A. and Christie M.J. Mu and delta receptors belong to a family of receptors that are coupled to potassium channels. Proceedings of the National Academy of Sciences; USA. (1987). 84: 5487-5491.

Nylander I., Vlaskovska M., Terenius L. Brain dynorphin and enkephalin systems in Fischer and Lewis rats: effects of morphine tolerance and withdrawal. Brain Res. (1995). 683: 25-35.

240 BIBLIOGRAFÍA

Ohsawa M., Mizoguchi H., Narita M., Chu M., Nagase H., Tseng L.F. Differential mechanisms mediating descending pain controls for antinociception induced by supraspinally administered endomorphin-1 and endomorphin-2 in the mouse. J Pharmacol Exp Ther. (2000). 294: 1106-1111.

Ohsawa M., Mizoguchi H., Narita M., Nagase H., Kampine J.P., Tseng L.F. Differential antinociception induced by spinally administered endomorphin-1 and endomorphin-2 in the mouse. J Pharmacol Exp Ther. (2001). 298: 592-297.

Olive M.F., Bertolucci M., Evans C.J., Maidment N.T. Microdialysis reveals a morphine-induced increase in pallidal opioid peptide release. NeuroReport. (1995). 6: 1093-1096.

Oliverio A., Castellano C., Eleftheriou B.E. Morphine sensitivity and tolerance: a genetic investigation in the mouse. Psychopharmacologia. (1975). 42: 219-224.

Olson G.A., Olson R.D., Kastin A.J. Endogenous opiates: 1996. Peptides. (1997). 18: 1651-1688.

Omiya Y., Goto K., Ishige A., Komatsu Y. Changes in analgesia-producing mechanism of repeated cold stress loading in mice. Pharmacol Biochem Behav. (2000). 65: 261-266.

Ossipov M.H., Kovelowski C.J., Nichols M.L., Hruby V.J., Porreca F. Characterization of supraspinal antinociceptive actions of opioid delta agonists in the rat. Pain. (1995) 62: 287-293.

Ossipov M.H., Kovelowski C.J., Porreca F. The increase in morphine antinociceptive potency produced by carrageenan-induced hindpaw inflammation is blocked by naltrindole, a selective delta-. Neurosci Lett. (1995b). 184: 173-176.

Ossipov M.H., Kovelowski C.J., Vanderah T., Porreca F. Naltrindole, an opioid delta antagonist, blocks the enhancement of morphine-antinociception induced by a CCKB antagonist in the rat. Neurosci Lett. (1994). 181: 9-12.

241 BIBLIOGRAFÍA

Ossipov M.H., Kovelowski C.J., Wheeler-Aceto H., Cowan A., Hunter J.C., Lai J., Malan T.P. Jr., Porreca F. Opioid antagonists and antisera to endogenous opioids increase the nociceptive response to formalin: demonstration of an opioid kappa and delta inhibitory tone. J Pharmacol Exp Ther. (1996). 277: 784-788.

Ossipov M.H., Lai J., Vanderah T.W., Porreca F. Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sci. (2003). 73: 783-800.

Ozaki S., Narita M., Narita M., Iino M., Miyoshi K., Suzuki T. Suppression of the morphine-induced rewarding effect and G-protein activation in the lower midbrain following nerve injury in the mouse: involvement of G-protein- coupled receptor kinase 2. Neuroscience. (2003). 116: 89-97

Pak Y., Kouvelas A., Scheideler M.A., Rasmussen J., O'Dowd B.F., George S.R. Agonist-induced functional desensitization of the mu-opioid receptor is mediated by loss of membrane receptors rather than uncoupling from G protein. Mol Pharmacol. (1996). 50: 1214-1222.

Pan Y.X., Xu J., Bolan E., Abbadie C., Chang E., Zckerman A., Rossi G., Pasternak G.W. Identification and characterization of three new alternatively spliced mu- opioid receptor isoforms. Molecular Pharmacology (1999). 56: 396-403.

Pan Y.X., Xu J., Mahurter L., Bolan E., Xu M., Pasternak G.W. Generation of the mu opioid receptor (MOR-1) protein by three new splice variants of the Oprm gene. Proc Natl Acad Sci U S A. (2001). 98: 4084-4089.

Panerai A.E., Martini A., Sacerdote P., Mantegazza P. Kappa-receptor antagonist reverse 'non-opioid' stress-induced analgesia. Brain Res. (1984). 304: 153-156.

Panocka I., Marek P., Sadowski B. Differentiation of neurochemical basis of stress- induced analgesia in mice by selective breeding. Brain Res. (1986). 397: 156- 160.

242 BIBLIOGRAFÍA

Parenti M., Tirone F., Giagnoni G., Pecora N., Parolaro D. Pertussis toxin inhibits the antinociceptive action of morphine in the rat. Eur J Pharmacol. (1986). 124: 357-359.

Paronis C.A., Holtzman S.G. Development of tolerance to the analgesic activity of mu agonists after continuous infusion of morphine, meperidine or fentanyl in rats. J Pharmacol Exp Ther. (1992). 262: 1-9.

Pasternak G.W. Hih and low affinity opioide binding sites: relationship to mu and delta sites. Life Sciences. (1982). 31: 1303-1306.

Pavlovic Z.W., Bodnar R.J. Opioid supraspinal analgesic synergy between the amygdala and periaqueductal gray in rats. Brain Res. (1998). 779: 158-169.

Pazos A., Florez J. Interaction of naloxone with mu- and delta-opioid agonists on the respiration of rats. Eur J Pharmacol. (1983). 87: 309-314.

Pazos A., Tristan C., Florez J. A comparative study of the respiratory depressant and analgesic effects of , a kappa-agonist. Life Sci. (1983). 33 (Suppl 1): 579-581.

Pei G., Kieffer B.L., Lefkowitz R.J., Freedman N.J. Agonist-dependent phosphorylation of the mouse delta-opioid receptor: involvement of G protein- coupled receptor kinases but not protein kinase C. Mol Pharmacol. (1995). 48: 173-177.

Pert C.B., Snyder S.H. Opiate receptor: demonstration in nervous tissue. Science. (1973). 179: 1011-1014.

Phillips A.G., LePiane F.G. Reinforcing effects of morphine microinjection into the ventral tegmental area. Pharmacol Biochem Behav. (1980). 12: 965-968.

Pick C.G., Cheng J., Paul D., Pasternak G.W. Genetic influences in opioid analgesic sensitivity in mice. Brain Res. (1991). 566: 295-298.

243 BIBLIOGRAFÍA

Pick C.G., Roques B., Gacel G., Pasternak G.W. Supraspinal mu 2-opioid receptors mediate spinal/supraspinal morphine synergy. Eur J Pharmacol. (1992). 220: 275-277.

Plesan A., Hoffmann O., Xu X.J., Wiesenfeld-Hallin Z. Genetic differences in the antinociceptive effect of morphine and its potentiation by dextromethorphan in rats. Neurosci Lett. (1999). 263: 53-56.

Plone M.A., Emerich D.F., Lindner M.D. Individual differences in the hotplate test and effects of habituation on sensitivity to morphine. Pain. (1996). 66: 265-270.

Polakiewicz R.D., Schieferl S.M., Dorner L.F., Kansra V., Comb M.J. A mitogen- activated protein kinase pathway is required for mu-opioid receptor desensitization. J Biol Chem. (1998). 273: 12402-12406.

Porreca F., Heyman J.S., Mosberg H.I., Omnaas J.R., Vaught J.L. Role of mu and

delta receptors in the supraspinal and spinal analgesic effects of [D-Pen2, D-

Pen5]enkephalin in the mouse. J Pharmacol Exp Ther. (1987). 241: 393-400.

Porreca F., Jiang Q., Tallarida R.J. Modulation of morphine antinociception by

peripheral [Leu5]enkephalin: a synergistic interaction. Eur J Pharmacol. (1990). 179: 463-468.

Portoghese P.S., Lunzer M.M. Identity of the putative delta1-opioid receptor as a delta-kappa heteromer in the mouse spinal cord. Eur J Pharmacol. (2003). 467: 233-234.

Portoghese P.S., Sultana M., Takemori A.E. Naltrindole, a highly selective and potent non-peptide delta opioid receptor antagonist. Eur J Pharmacol. (1988). 146(1): 185-186.

Pugh G. Jr., Mason DJ. Jr., Combs V., Welch S.P. Involvement of in the antinociceptive effects of the cannabinoid CP55,940 in the spinal cord. J Pharmacol Exp Ther. (1997). 281: 730-737.

244 BIBLIOGRAFÍA

Pugh G. Jr., Smith P.B., Dombrowski D.S., Welch S.P. The role of endogenous opioids in enhancing the antinociception produced by the combination of delta 9- and morphine in the spinal cord. J Pharmacol Exp Ther. (1996). 279: 608-616.

Quock R.M., Burkey T.H., Varga E., Hosohata Y., Hosohata K., Cowell S.M., Slate C.A., Ehlert F.J., Roeske W.R., Yamamura H.I. The delta-opioid receptor: molecular pharmacology, signal transduction, and the determination of drug efficacy. Pharmacol Rev. (1999). 51: 503-532.

Raffa R.B., Martinez R.P., Porreca F. Lack of antinociceptive efficacy of

intracerebroventricular [D-Ala2,Glu4]deltorphin, but not [D-Pen2,D-

Pen5]enkephalin, in the mu-opioid receptor deficient CXBK mouse. Eur J Pharmacol. (1992). 216: 453-456.

Ramarao P., Jablonski H.I. Jr., Rehder K.R., Bhargava H.N. Effect of kappa-opioid receptor agonists on morphine analgesia in morphine-naive and morphine- tolerant rats. Eur J Pharmacol. (1988). 156: 239-246.

Rasmussen K., Beitner-Johnson D.B., Krystal J.H., Aghajanian G.K., Nestler E.J. Opiate withdrawal and the rat locus coeruleus: behavioral, electrophysiological, and biochemical correlates. J Neurosci. (1990). 10: 2308-2317.

Raynor K., Kong H., Mestek A., Bye L.S., Tian M., Liu J., Yu L., Reisine T. Characterization of the cloned human mu opioid receptor. J Pharmacol Exp Ther. (1995). 272: 423-428.

Reisine T., Pasternak G.W. Opioid analgesics and antagonist. En Hardman JG, Limbird LE, Molinoff PB, Ruddon RW y Goodman GA (Eds.). The pharmacological basis of therapeutics. Goodman and Gilman´s (1996).

Remmers A.E., Clark M.J., Mansour A., Akil H., Woods J.H., Medzihradsky F. Opioid efficacy in a C6 glioma cell line stably expressing the human kappa opioid receptor. J Pharmacol Exp Ther. (1999). 288: 827-833.

245 BIBLIOGRAFÍA

Richardson A., Demoliou-Mason C., Barnard E.A. Guanine nucleotide-binding protein-coupled and -uncoupled states of opioid receptors and their relevance to the determination of subtypes. Proc Natl Acad Sci U S A. (1992). 89: 10198- 10202.

Robinson T.E., Berridge K.C. Incentive-sensitization and addiction. Addiction. (2001). 96: 103-14.

Robson L.E., Gillan M.G., Kosterlitz H.W. Species differences in the concentrations and distributions of opioid binding sites. Eur J Pharmacol. (1985). 112: 65-71.

Roerig S.C. Decreased spinal morphine/clonidine antinociceptive synergism in morphine-tolerant mice. Life Sci. (1995). 56: PL115-122.

Roerig S.C., Fujimoto JM. Multiplicative interaction between intrathecally and intracerebroventricularly administered mu opioid agonists but limited interactions between delta and kappa agonists for antinociception in mice. J Pharmacol Exp Ther. (1989). 249: 762-768.

Roques B.P., Noble F., Fournié-Zaluski M.C. Peptidos opioides y analgesia. En: Stein C. (Eds.). Opioides en el control del dolor. Aspectos básicos y clínicos. Masson, Barcelona. (2001): 21-44.

Rosecrans J.A., Robinson S.E., Johnson J.H., Mokler D.J., Hong J.S. Neuroendocrine, biogenic amine and behavioral responsiveness to a repeated foot-shock-induced analgesia (FSIA) stressor in Sprague-Dawley (CD) and Fischer-344 (CDF) rats. Brain Res. (1986). 382: 71-80.

Rossi G.C., Brown G.P., Leventhal L., Yang K., Pasternak G.W. Novel receptor mechanisms for heroin and morphine-6 beta-glucuronide analgesia. Neurosci Lett. (1996). 216: 1-4.

Rossi G.C., Pan Y.X., Brown G.P., Pasternak G.W. Antisense Mapping the MOR-1 opioid receptor: evidence for alternative splicing and a novel morphine-6 beta-

246 BIBLIOGRAFÍA

glucuronide receptor. Federation of European Biological Societies Letters. (1995b). 369: 192-196.

Rossi G.C., Pasternak G.W., Bodnar R.J. Synergistic brainstem interactions for morphine analgesia. Brain Res. (1993). 624: 171-180.

Rossi G.C., Pasternak G.W., Bodnar R.J. Mu and delta opioid synergy between the periaqueductal gray and the rostro-ventral medulla. Brain Res. (1994). 665: 85- 93.

Rossi G.C., Standifer K.M., Pasternak G.W. Differential blockade of morphine and morphine-6 beta-glucuronide analgesia by antisense oligodeoxynucleotides directed against MOR-1 and G-protein alpha subunits in rats. Neurosci Lett. (1995). 198: 99-102.

Rothman R.B., Bykov V., Long J.B., Brady L.S., Jacobson A.E., Rice K.C., Holaday J.W. Chronic administration of morphine and naltrexone up-regulate mu-opioid

3 binding sites labeled by [ H][D-Ala2,MePhe4,Gly-ol5]enkephalin: further evidence for two mu-binding sites. Eur J Pharmacol. (1989). 160: 71-82.

Rothman R.B., Westfall T.C. Morphine allosterically modulates the binding of [3H]leucine enkephalin to a particulate fraction of rat brain. Mol Pharmacol. (1982a). 21: 538-547.

Rothman R.B., Westfall T.C. Allosteric coupling between morphine and enkephalin receptors in vitro. Mol Pharmacol. (1982b). 21: 548-57.

Rubinstein M., Mogil J.S., Japon M., Chan E.C., Allen R.G., Low M.J. Absence of opioid stress-induced analgesia in mice lacking beta-endorphin by site-directed mutagenesis. Proc Natl Acad Sci U S A. (1996). 93: 3995-4000.

Rusovici D.E., Negus S.S., Mello N.K., Bidlack J.M. Kappa-opioid receptors are differentially labeled by arylacetamides and benzomorphans. Eur J Pharmacol. (2004). 485: 119-125.

247 BIBLIOGRAFÍA

Sakurada S., Hayashi T., Yuhki M., Orito T., Zadina J.E., Kastin A.J., Fujimura T., Murayama K., Sakurada C., Sakurada T., Narita M., Suzuki T., Tan-no K., Tseng L.F. Differential antinociceptive effects induced by intrathecally administered endomorphin-1 and endomorphin-2 in the mouse. Eur J Pharmacol. (2001). 427: 203-210.

Samama P., Cotecchia S., Costa T., Lefkowitz R.J. A mutation-induced activated state of the beta 2-adrenergic receptor. Extending the ternary complex model. J Biol Chem. (1993). 268: 4625-4636.

Sanchez-Blazquez P., Garzon J. Pertussis toxin differentially reduces the efficacy of opioids to produce supraspinal analgesia in the mouse. Eur J Pharmacol. (1988). 152: 357-361.

Sarton E., Teppema L., Dahan A. Sex differences in morphine-induced ventilatory depression reside within the peripheral chemoreflex loop. Anesthesiology. (1999). 90: 1329-1338.

Satoh M., Minami M. Molecular pharmacology of the opioid receptors. Pharmacology and Therapeutics. (1995). 68: 343-364.

Schmidt B.L., Tambeli C.H., Levine J.D., Gear R.W. Mu/delta Cooperativity and opposing kappa-opioid effects in nucleus accumbens-mediated antinociception in the rat. Eur J Neurosci. (2002). 15: 861-868.

Schnoll S.H., Weaver M.F. Addiction and pain. Am J Addict. (2003). 12 (Suppl 2): S27-35.

Schomburg E.D., Restrictions on the interpretation of spinal reflex modulation in pain and analgesia research., Pain Forum. (1997). 6: 101.

Schroeder J.E., Fischbach P.S., Zheng D., McCleskey E.W. Activation of mu opioid receptors inhibits transient high- and low-threshold Ca2+ currents, but spares a sustained current. Neuron. (1991). 6: 13-20.

248 BIBLIOGRAFÍA

Schuller A.G., King M.A., Zhang J., Bolan E., Pan Y.X., Morgan D.J., Chang A., Czick M.E., Unterwald E.M., Pasternak G.W., Pintar J.E. Retention of heroin and morphine-6 beta-glucuronide analgesia in a new line of mice lacking exon 1 of MOR-1. Nat Neurosci. (1999). 2: 151-156.

Selley D.E., Liu Q., Childers S.R. Signal transduction correlates of mu opioid agonist intrinsic efficacy: receptor-stimulated [35S]GTP gamma S binding in mMOR- CHO cells and rat thalamus. J Pharmacol Exp Ther. (1998). 285: 496-505

Selley D.E., Nestler E.J., Breivogel C.S., Childers S.R. Opioid receptor-coupled G- proteins in rat locus coeruleus membranes: decrease in activity after chronic morphine treatment. Brain Res. (1997). 746: 10-18.

Selley D.E, Sim L.J., Xiao R., Liu Q., Childers S.R. Mu-Opioid receptor-stimulated guanosine-5'-O-(gamma-thio)-triphosphate binding in rat thalamus and cultured cell lines: signal transduction mechanisms underlying agonist efficacy. Mol Pharmacol. (1997b). 51: 87-96.

Selley D.E., Herbert J.T., Morgan D., Cook C.D., Picker M.J., Sim-Selley L.J. Effect of strain and sex on mu opioid receptor-mediated G-protein activation in rat brain. Brain Res Bull. (2003). 60: 201-208.

Semenova S., Kuzmin A., Zvartau E. Strain differences in the analgesic and reinforcing action of morphine in mice. Pharmacol Biochem Behav. (1995). 50: 17-21.

Shannon H.E., Lutz E.A. Comparison of the peripheral and central effects of the opioid agonists and morphine in the formalin test in rats. Neuropharmacology. (2002). 42: 253-261.

Sharma S.K., Klee W.A., Nirenberg M. Dual regulation of adenylate cyclase accounts for narcotic dependence and tolerance. Proc Natl Acad Sci U S A. (1975). 72: 3092-3096.

249 BIBLIOGRAFÍA

Sharma S.K., Nirenberg M., Klee W.A. Morphine receptors as regulators of adenylate cyclase activity. Proc Natl Acad Sci U S A. (1975). 72: 590-594.

Shimomura K., Kamata O., Ueki S., Ida S., Oguri K., Yoshimura H., Tsukamoto H. Analgesic effect of morphine glucuronides. Tohoku J. Exp. Med. (1971). 105: 45-52.

Shoaib M., Spanagel R., Stohr T., Shippenberg T.S. Strain differences in the rewarding and dopamine-releasing effects of morphine in rats. Psychopharmacology. (1995). 117: 240-247.

Sim L.J., Selley D.E., Childers S.R. In vitro autoradiography of receptor-activated G proteins in rat brain by agonist-stimulated guanylyl 5'-[gamma-[35S]thio]- triphosphate binding. Proc Natl Acad Sci U S A. (1995). 92: 7242-7246.

Sim L.J., Selley D.E., Dworkin S.I., Childers S.R. Effects of chronic morphine administration on mu opioid receptor-stimulated [35S]GTPgammaS autoradiography in rat brain. J Neurosci. (1996). 16: 2684-2692.

Simon E.J., Hiller J.M., Edelman I. Stereospecific binding of the potent narcotic analgesic [3H] to rat-brain homogenate. Proc Natl Acad Sci U S A. (1973). 70: 1947-1949.

Simonin F., Valverde O., Smadja C., Slowe S., Kitchen I., Dierich A., Le Meur M., Roques B.P., Maldonado R., Kieffer B.L. Disruption of the kappa-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective kappa-agonist U-50,488H and attenuates morphine withdrawal. EMBO J. (1998). 17: 886-897.

Smith F.L., Cichewicz D., Martin Z.L., Welch S.P. The enhancement of morphine antinociception in mice by delta9-tetrahydrocannabinol. Pharmacol Biochem Behav. (1998). 60: 559-566.

Smith A.P., Lee N.M. Opioid receptor interactions: local and nonlocal, symmetric and asymmetric, physical and functional. Life Sci. (2003). 73: 1873-1893.

250 BIBLIOGRAFÍA

Smith M.T., Watt J.A., Cramond T. Morphine-3-glucuronide--a potent antagonist of morphine analgesia. Life Sci. (1990). 47: 579-585.

Snyder S.H., Pasternak G.W., Pert C. En: Iversen L.L., Iverson S.D., Zinder S.H. (Eds.). Handbook of psychopharmacology. Plenum, New York. (1973). 5: 329- 360.

Sofuoglu M., Portoghese P.S., Takemori A.E. Cross-tolerance studies in spinal cord of beta->FNA-treated mice provides further evidence for delta opioide receptor subtypes. Life Sci. (1991a). 49: P1153-1156.

Sofuoglu M., Portoghese P.S., Takemori A.E. Differential antagonism of delta opioide agonists by naltrindole and its benzofuran analog (NTB) in mice: evidence for delta opioide subtypes. J Pharmacol Exp Ther. (1991b). 257: 676- 680.

Sora I., Takahashi N., Funada M., Ujike H., Revay R.S., Donovan D.M., Miner L.L., Uhl G.R. Opiate receptor knockout mice define mu receptor roles in endogenous nociceptive responses and morphine-induced analgesia. Proc Natl Acad Sci U S A. (1997). 94: 1544-1549.

Spanagel R., Herz A., Shippenberg T.S. The effects of opioid peptides on dopamine release in the nucleus accumbens: an in vivo microdialysis study. J Neurochem. (1990). 55: 1734-1740.

Stachura Z., Herman Z.S. The influence of the kappa agonist- (U62066E) on the analgesic activity of some opioids at the spinal level. Pol J Pharmacol. (1994). 46: 37-41.

Stanfa L.C., Dickenson A.H. Cholecystokinin as a factor in the enhanced potency of spinal morphine following carrageenin inflammation. Br J Pharmacol. (1993). 108: 967-973.

Stefano G.B., Hartman A., Bilfinger T.V., Magazine H.I., Liu Y., Casares F., Goligorsky M.S. Presence of the mu3 opiate receptor in endotelial cells.

251 BIBLIOGRAFÍA

Coupling to production and vasodilation. Journal of Biological Chemistry. (1995). 270: 30290-30293.

Stein C. Peripheral mechanisms of opioid analgesia. Anesth Analg. (1993). 76: 182- 191.

Stein C., Cabot P.J., Schafer M. Mecanismos supraespinales de la analgesia opioide. En: Opioides en el control del dolor. Aspectos básicos y clínicos. Dir. Christoph Stein. Ed. Masson, 2001 Barcelona. p. 93-104.

Sternberg E.M., Young W.S. 3rd., Bernardini R., Calogero A.E., Chrousos G.P., Gold P.W., Wilder R.L. A central nervous system defect in biosynthesis of corticotropin-releasing hormone is associated with susceptibility to streptococcal cell wall-induced arthritis in Lewis rats. Proc Natl Acad Sci U S A. (1989). 86: 4771-4775.

Stewart J., Badiani A. Tolerance and sensitization to the behavioral effects of drugs. Behav Pharmacol. (1993). 4: 289-312.

Stinus L., Allard M., Gold L., Simonnet G. Changes in CNS neuropeptide FF-like material, pain sensitivity, and opiate dependence following chronic morphine treatment. Peptides. (1995). 16: 1235-1241.

Stoffel E.C., Ulibarri C.M., Craft R.M. Gonadal steroid hormone modulation of nociception, morphine antinociception and reproductive indices in male and female rats. Pain. (2003). 103: 285-302.

Su Y.F., McNutt R.W., Chang K.J. Delta-opioid ligands reverse -induced respiratory depression but not antinociception. J Pharmacol Exp Ther. (1998). 287: 815-823.

Surprenant A., Shen K.Z., North R.A., Tatsumi H. Inhibition of calcium currents by noradrenaline, somatostatin and opioids in guinea-pig submucosal neurones. J Physiol. (1990). 431: 585-608.

252 BIBLIOGRAFÍA

Sutters K.A., Miaskowski C., Taiwo Y.O., Levine J.D. Analgesic synergy and improved motor function produced by combinations of mu-delta- and mu- kappa-opioids. Brain Res. (1990). 530: 290-294.

Suzuki T., Ikeda H., Tsuji M., Misawa M., Narita M., Tseng L.F. Antisense oligodeoxynucleotide to delta opioid receptors attenuates morphine dependence in mice. Life Sci. (1997). 61: PL 165-170.

Suzuki T., Kishimoto Y., Ozaki S., Narita M. Mechanism of opioid dependence and interaction between opioid receptors. Eur J Pain. (2001). 5 (Suppl A): 63-65.

Suzuki T., Narita M., Takahashi Y., Misawa M., Nagase H. Effects of nor- on the development of analgesic tolerance to and physical dependence on morphine. Eur J Pharmacol. (1992). 213: 91-97.

Suzuki T., Tsuji M., Mori T., Misawa M., Nagase H. Involvement of delta 1 and delta 2 opioid receptor subtypes in the development of physical dependence on morphine in mice. Pharmacol Biochem Behav. (1997b). 57: 293-299.

Suzuki T., Yoshiike M., Mizoguchi H., Kamei J., Misawa M., Nagase H. Blockade of delta-opioid receptors prevents morphine-induced place preference in mice. Jpn J Pharmacol. (1994). 66: 131-137.

Svoboda K.R., Lupica C.R. Opioid inhibition of hippocampal interneurons via modulation of potassium and hyperpolarization-activated cation (Ih) currents. J Neurosci. (1998). 18: 7084-7098.

Szeto H.H., Soong Y., Wu D., Qian X., Zhao G.M. Endogenous opioid peptides contribute to antinociceptive potency of intrathecal [Dmt1]DALDA. J Pharmacol Exp Ther. (2003). 305: 696-702.

Takemori A.E., Ho B.Y., Naeseth J.S., Portoghese P.S. Nor-binaltorphimine, a highly selective kappa-opioid antagonist in analgesic and receptor binding assays. J Pharmacol Exp Ther. (1988). 246: 255-258.

253 BIBLIOGRAFÍA

Takemori A.E., Portoghese P.S. Evidence for the interaction of morphine with kappa and delta opioid receptors to induce analgesia in beta-funaltrexamine-treated mice. J Pharmacol Exp Ther. (1987). 243: 91-94.

Tan P.P., Chen J.C., Li J.Y., Liang K.W., Wong C.H., Huang E.Y. Modulation of naloxone-precipitated morphine withdrawal syndromes in rats by neuropeptide FF analogs. Peptides. (1999). 20: 1211-1217.

Tang N.M., Dong H.W., Wang X.M., Tsui Z.C., Han J.S. Cholecystokinin antisense RNA increases the analgesic effect induced by electroacupuncture or low dose morphine: conversion of low responder rats into high responders. Pain. (1997). 71: 71-80.

Tao P.L., Han K.F., Wang S.D., Lue W.M., Elde R., Law P.Y., Loh H.H. Immunohistochemical evidence of down-regulation of mu-opioid receptor after chronic PL-017 in rats. Eur J Pharmacol. (1998). 344: 137-142.

Taylor D.A., Fleming W.W. Unifying perspectives of the mechanisms underlying the development of tolerance and physical dependence to opioids. J Pharmacol Exp Ther. (2001). 297: 11-18.

Terenius L. Characteristics of the "receptor" for narcotic analgesics in synaptic plasma membrane fraction from rat brain. Acta Pharmacol Toxicol. (1973). 33: 377-384.

Terman G.W., Morgan M.J., Liebeskind J.C. Opioid and non-opioid stress analgesia from cold water swim: importance of stress severity. Brain Res. (1986). 372: 167-171.

Thomas J.B., Herault X.M., Rothman R.B., Burgess J.P., Mascarella S.W., Xu H., Horel R.B., Dersch C.M., Carroll F.I. (+/-)-4-[(N-allyl-cis-3-methyl-4- piperidinyl)phenylamino]-N,N-diethylbenzamide displays selective binding for the delta opioid receptor. Bioorg Med Chem Lett. (1999). 9: 3053-3056.

254 BIBLIOGRAFÍA

Thornton S.R., Smith F.L. Characterization of neonatal rat fentanyl tolerance and dependence. J Pharmacol Exp Ther. (1997). 281: 514-521.

Tiberi M., Magnan J. Demonstration of the heterogeneity of the kappa-opioid receptors in guinea-pig cerebellum using selective and nonselective drugs. Eur J Pharmacol. (1990). 188: 379-389.

Tierney G., Carmody J., Jamieson D. Stress analgesia: the opioid analgesia of long swims suppresses the non-opioid analgesia induced by short swims in mice. Pain. (1991). 46: 89-95.

Tiseo P.J., Yaksh T.L. Dose-dependent antagonism of spinal opioid receptor agonists by naloxone and naltrindole: additional evidence for delta-opioid receptor subtypes in the rat. Eur J Pharmacol. (1993). 236: 89-96

Traynor J.R., Elliott J. Delta-Opioid receptor subtypes and cross-talk with mu- receptors. Trends Pharmacol Sci. (1993). 14: 84-86.

Traynor J.R., Hunter J.C., Rodriguez R.E., Hill R.G., Hughes J. Delta-opioid receptor binding sites in rodent spinal cord. Br J Pharmacol. (1990). 100: 319- 323.

Traynor J.R., Nahorski S.R. Modulation by mu-opioid agonists of guanosine-5'-O-(3- [35S]thio)triphosphate binding to membranes from human neuroblastoma SH- SY5Y cells. Mol Pharmacol. (1995). 47: 848-854.

Trouth C.O., Bada F.J., Pan Y., Holloway J.A., Millis R.M., Bernard D.G. Naloxone application to the ventrolateral medulla enhances the respiratory response to inspired carbon dioxide. Life Sci. (1991). 49: 193-200.

Trujillo K.A. Are NMDA receptors involved in opiate-induced neural and behavioral plasticity? A review of preclinical studies. Psychopharmacology. (2000). 151: 121-141.

255 BIBLIOGRAFÍA

Trujillo K.A. and Akil H. Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science. (1991). 251: 85-87.

Tseng L.F. Evidence for epsilon-opioid receptor-mediated beta-endorphin-induced analgesia. Trends Pharmacol Sci. (2001). 22: 623-630.

Tseng L.F., Collins K.A. Spinal involvement of both dynorphin A and Met- enkephalin in the antinociception induced by intracerebroventricularly administrered bremazocine but not morphine in the mouse. J Pharmacol Exp Ther. (1993). 266: 1430-1438.

Tseng L.F., Collins K.A. Antisense oligodeoxynucleotide to a delta-opioid receptor given intrathecally blocks i.c.v. administered beta-endorphin-induced antinociception in the mouse. Life Sci. (1994). 55: PL127-131.

Tseng L.F., Higgins M.J., Hong J.S., Hudson P.M., Fujimoto J.M. Release of immunoreactive Met-enkephalin from the spinal cord by intraventricular beta- endorphin but not morphine in anesthetized rats. Brain Res. (1985). 343: 60-69.

Tsuji M., Narita M., Mizoguchi H., Narita M., Ohsawa M., Kamei J., Nagase H., Takeda H., Matsumiya T., Tseng L.F. Region-dependent G-protein activation by mu-, delta 1- and delta 2-opioid receptor agonists in the brain: comparison between the midbrain and forebrain. Life Sci. (1999). 65: PL233-239.

Tsuji M., Yamazaki M., Takeda H., Matsumiya T., Nagase H., Tseng L.F., Narita M., Suzuki T. The novel kappa-opioid receptor agonist TRK-820 has no affect on the development of antinociceptive tolerance to morphine in mice. Eur J Pharmacol. (2000). 394: 91-95.

Tulunay F.C., Jen M.F., Chang J.K., Loh H.H., Lee N.M. Possible regulatory role of dynorphin on morphine- and beta-endorphin-induced analgesia. J Pharmacol Exp Ther. (1981). 219: 296-298.

Twycross R.G. Stong narcotic analgesics. Clin. Oncol. (1984). 3: 109-133.

256 BIBLIOGRAFÍA

Tzschentke T.M. Measuring reward with the conditioned place preference paradigm: a comprehensive review of drug effects, recent progress and new issues. Progress in Neurobiology. (1998). 56: 613-672.

Ueda H., Inoue M., Mizuno K. New approaches to study the development of morphine tolerance and dependence. Life Sci. (2003). 74: 313-320.

Ueda H., Inoue M., Takeshima H., Iwasawa Y. Enhanced spinal expression develops morphine tolerance and dependence. J Neurosci. (2000). 20: 7640-7647.

Ueda H., Miyamae T., Hayashi C., Watanabe S., Fukushima N., Sasaki Y., Iwamura T., Misu Y. Protein kinase C involvement in homologous desensitization of delta-opioid receptor coupled to Gi1-phospholipase C activation in Xenopus oocytes. J Neurosci. (1995). 15: 7485-7499.

Ueda H., Yamaguchi T., Tokuyama S., Inoue M., Nishi M., Takeshima H. Partial loss of tolerance liability to morphine analgesia in mice lacking the nociceptin receptor gene. Neurosci Lett. (1997). 237: 136-138.

Vaccarino F.J., Bloom F.E., Koob G.F. Blockade of nucleus accumbens opiate receptors attenuates intravenous heroin reward in the rat. Psychopharmacology. (1985). 86: 37-42.

Vaccarino A.L., Couret L.C. Jr. Relationship between hypothalamic-pituitary-adrenal activity and blockade of tolerance to morphine analgesia by pain: a strain comparison. Pain. (1995). 63: 385-389.

Vaccarino A.L., Marek P., Kest B., Ben-Eliyahu S., Couret L.C. Jr., Kao B., Liebeskind J.C. Morphine fails to produce tolerance when administered in the presence of formalin pain in rats. Brain Res. (1993). 627: 287-290.

Valverde O., Fournie-Zaluski M.C., Roques B.P., Maldonado R. The CCKB antagonist PD-134,308 facilitates rewarding effects of endogenous enkephalins

257 BIBLIOGRAFÍA

but does not induce place preference in rats. Psychopharmacology. (1996). 123: 119-126.

Valverde O., Maldonado R., Fournie-Zaluski M.C., Roques B.P. Cholecystokinin B antagonists strongly potentiate antinociception mediated by endogenous enkephalins. J Pharmacol Exp Ther. (1994). 270: 77-88.

Vanderah T.W., Bernstein R.N., Yamamura H.I., Hruby V.J., Porreca F. Enhancement of morphine antinociception by a CCKB antagonist in mice is mediated via opioid delta receptors. J Pharmacol Exp Ther. (1996). 278: 212- 219.

Vanderah T.W., Wild K.D., Takemori A.E., Sultana M., Portoghese P.S., Bowen W.D., Hruby V.J., Mosberg H.I., Porreca F. Modulation of morphine antinociception by swim-stress in the mouse: involvement of supraspinal opioid delta-2 receptors. J Pharmacol Exp Ther. (1993). 267: 449-455.

Vanderah T.W., Wild K.D., Takemori A.E., Sultana M., Portoghese P.S., Bowen W.D., Mosberg H.I., Porreca F. Mediation of swim-stress antinociception by the opioid delta 2 receptor in the mouse. J Pharmacol Exp Ther. (1992). 262: 190-197.

Vanderschuren L.J., De Vries T.J., Wardeh G., Hogenboom F.A., Schoffelmeer A.N. A single exposure to morphine induces long-lasting behavioural and neurochemical sensitization in rats. Eur J Neurosci. (2001). 14: 1533-1538.

Vaswani K.K., Richard C.W., Tejwani I.I.I. and G.A. Cold Swim Stress-Induced Changes in the Levels of Opioid Peptides in the Rat CNS and Peripheral Tissues. Pharmacol. Biochem. Behav. (1988). 29: 163-168.

Vaught J.L., Takemori A.E. Differential effects of leucine and methionine enkephalin on morphine-induced analgesia, acute tolerance and dependence. J Pharmacol Exp Ther. (1979). 208: 86-90

258 BIBLIOGRAFÍA

Vaughn L.K., Knapp R.J., Toth G., Wan Y.P., Hruby V.J., Yamamura H.I. A high

3 affinity, highly selective ligand for the delta opioid receptor: [ H]-[D-Pen2, pCl-

Phe4, d-Pen5]enkephalin. Life Sci. (1989). 45: 1001-1008.

Verborgh C., Meert T.F. The effects of intravenous naltrindole and naltrindole 5'- isothiocyanate on -induced respiratory depression and antinociception in rats. Pharmacol Biochem Behav. (1999). 63: 175-183.

Von Voigtlander P.F., Lahti R.A., Ludens J.H. U-50,488: a selective and structurally novel non-Mu (kappa) opioid agonist. J Pharmacol Exp Ther. (1983). 224: 7- 12.

Walker J.M., Thompson L.A., Frascella J., Friederich M.W. Opposite effects of mu and kappa opiates on the firing-rate of dopamine cells in the substantia nigra of the rat. Eur J Pharmacol. (1987). 134: 53-59.

Wang X.J., Han J.S. Modification by cholecystokinin octapeptide of the binding of mu-, delta-, and kappa-opioid receptors. J Neurochem. (1990). 55: 1379-1382.

Wang J.B., Imai Y., Eppler C.M., Gregor P., Spivak C.E., Uhl G.R. Mu opiate receptor: cDNA cloning and expression. Proc Natl Acad Sci U S A. (1993). 90: 10230-10234.

Wang Z., Gardell L.R., Ossipov M.H., Vanderah T.W., Brennan M.B., Hochgeschwender U., Hruby V.J., Malan T.P. Jr., Lai J., Porreca F. Pronociceptive actions of dynorphin maintain chronic neuropathic pain. J Neurosci. (2001). 21: 1779-1786.

Wang J., Ren M., Han J. Mobilization of calcium from intracellular stores as one of the mechanisms underlying the antiopioid effect of cholecystokinin octapeptide. Peptides. (1992). 13: 947-951.

Ward S.J., Portoghese P.S., Takemori A.E. Improved assays for the assessment of kappa- and delta-properties of opioid ligands. Eur J Pharmacol. (1982). 85: 163-170.

259 BIBLIOGRAFÍA

Watson G.S., Sufka K.J., Coderre T.J. Optimal scoring strategies and weights for the formalin test in rats. Pain. (1997). 70: 53-58.

Way E.L., Loh H.H., Shen F.H. Simultaneous quantitative assessment of morphine tolerance and physical dependence. J. Pharmacol. Exp. Ther. (1969). 167: 1-8.

Welch S.P., Stevens D.L. Antinociceptive activity of intrathecally administered cannabinoids alone, and in combination with morphine, in mice. J Pharmacol Exp Ther. (1992). 262: 10-18.

Werling L.L., Frattali A., Portoghese P.S., Takemori A.E., Cox B.M. Kappa receptor regulation of dopamine release from striatum and cortex of rats and guinea pigs. J Pharmacol Exp Ther. (1988). 246: 282-286.

Wiesenfeld-Hallin Z., Xu X. Interacciones opioides-no opioides. En Stein C. (Eds.). Opioides en el control del dolor. Aspectos básicos y clínicos. Masson, Barcelona. (2001): 127-138.

Wigdor S., Wilcox G.L. Central and systemic morphine-induced antinociception in mice: contribution of descending serotonergic and noradrenergic pathways. J Pharmacol Exp Ther. (1987). 242: 90-95.

Wild K.D., McCormick J., Bilsky E.J., Vanderah T., McNutt R.W., Chang K.J., Porreca F. Antinociceptive actions of BW373U86 in the mouse. J Pharmacol Exp Ther. (1993). 267: 858-865.

Willer J.C., Albe-Fessard D. Eledctrophysiological evidence for a release of endogenous opiates in stress-induced “analgesia” in man. Brain Res. (1980). 198: 419-426.

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

Willis W.D., Westlund K.N. Neuroanatomy of the pain system and of the pathways that modulate pain. J Clin Neurophysiol. (1997). 14: 2-31.

260 BIBLIOGRAFÍA

Wilson S.G., Chesler E.J., Hain H., Rankin A.J., Schwarz J.Z., Call S.B., Murray M.R., West E.E., Teuscher C., Rodriguez-Zas S., Belknap J.K., Mogil J.S. Identification of quantitative trait loci for chemical/inflammatory nociception in mice. Pain. (2002). 96: 385-391.

Wilson S.G., Smith S.B., Chesler E.J., Melton K.A., Haas J.J., Mitton B., Strasburg K., Hubert L., Rodriguez-Zas S.L., Mogil J.S. The heritability of antinociception: common pharmacogenetic mediation of five neurochemically distinct analgesics. J Pharmacol Exp Ther. (2003). 304: 547-559.

Wise R.A. Neurobiology of addiction. Curr Opin Neurobiol. (1996). 6: 243-251.

Wollemann M., Benyhe S., Simon J. The kappa-opioid receptor: evidence for the different subtypes. Life Sci. (1993). 52: 599-611.

Woolfe G., MacDonald A.L. The evaluation of the analgesic action of pethidine hydrochloride (Demerol). J. Pharmacol. Exp. Ther. (1944). 80: 300-307.

Woolfolk D.R., Holtzman S.G. Rat strain differences in the potentiation of morphine-induced analgesia by stress. Pharmacol Biochem Behav. (1995). 51: 699-703.

Wu H.E., Hung K.C., Mizoguchi H., Nagase H., Tseng L.F. Roles of endogenous opioid peptides in modulation of nocifensive response to formalin. J Pharmacol Exp Ther. (2002). 300: 647-654.

Wuster M., Schulz R., Herz A. Specificity of opioids towards the mu-, delta- and epsilon-opiate receptors. Neurosci Lett. (1979). 15: 193-198.

Xu M., Kontinen V.K., Panula P., Kalso E. Effects of (1DMe)NPYF, a synthetic neuropeptide FF analogue, in different pain models. Peptides. (1999). 20: 1071- 1077.

Xu M., Kontinen V.K., Panula P., Kalso E. Role of the delta-opioid receptor in (1DMe)NPYF mediated antinociception. Peptides. (2001). 22: 33-38.

261 BIBLIOGRAFÍA

Xu H., Smolens I., Gintzler A.R. Opioids can enhance and inhibit the electrically evoked release of methionine-enkephalin. Brain Res. (1989). 504: 36-42.

Yabaluri N., Medzihradsky F. Down-regulation of mu-opioid receptor by full but not partial agonists is independent of G protein coupling. Mol Pharmacol. (1997). 52: 896-902.

Yaksh T.L. Pharmacology and mechanisms of opioid analgesic activity. Acta Anaesthesiol Scand. (1997). 41(1 Pt 2) :94-111.

Yaksh T.L., Al-Rodhan N.R., Jensen T.S. Sites of action of opiates in production of analgesia. Prog Brain Res. (1988). 77: 371-394.

Yamamura M.S., Horvath R., Toth G., Otvos F., Malatynska E., Knapp R.J., Porreca F., Hruby V.J., Yamamura H.I. Characterization of [3H]naltrindole binding to delta opioid receptors in rat brain. Life Sci. (1992). 50: PL119-124.

Yang H.Y., Fratta W., Majane E.A., Costa E. Isolation, sequencing, synthesis, and pharmacological characterization of two brain neuropeptides that modulate the action of morphine. Proc Natl Acad Sci U S A. (1985). 82: 7757-7761.

Yeung J.C., Rudy T.A. Multiplicative interaction between narcotic agonisms expressed at spinal and supraspinal sites of antinociceptive action as revealed by concurrent intrathecal and intracerebroventricular injections of morphine. J Pharmacol Exp Ther. (1980). 215: 633-642.

You Z.B., Herrera-Marschitz M., Nylander I., Goiny M., Kehr J., Ungerstedt U., Terenius L. Effect of morphine on dynorphin B and GABA release in the basal ganglia of rats. Brain Res. (1996). 710: 241-248.

Zadina J.E., Chang S.L., Ge L.J., Kastin A.J. Mu opiate receptor down-regulation by morphine and up-regulation by naloxone in SH-SY5Y human neuroblastoma cells. J Pharmacol Exp Ther. (1993). 265: 254-262.

262 BIBLIOGRAFÍA

Zadina J.E., Hackler L., Ge L.J., Kastin A.J. A potent and selective endogenous agonist for the mu-opiate receptor. Nature. (1997). 386: 499-502.

Zagon I.S., Gibo D.M., McLaughlin P.J. Zeta (ζ), a growth-related opioid receptor in developing rat cerebellum: identification and characterization. Brain Res. (1991). 551: 28-35.

Zeng F.Y., Wess J. Identification and molecular characterization of m3 muscarinic receptor dimmers. Journal of Biological Chemistry. (1999). 274: 19487-19497.

Zhang J., Ferguson S.S., Barak L.S., Bodduluri S.R., Laporte S.A., Law P.Y., Caron M.G. Role for G protein-coupled receptor kinase in agonist-specific regulation of mu-opioid receptor responsiveness. Proc Natl Acad Sci U S A. (1998). 95: 7157-7162.

Zhao C.S., Tao Y.X., Tall J.M., Donovan D.M., Meyer R.A., Raja S.N. Role of mu- opioid receptors in formalin-induced pain behavior in mice. Exp Neurol. (2003). 184: 839-845.

Zhu Y., King M.A., Schuller A.G., Nitsche J.F., Reidl M., Elde R.P., Unterwald E., Pasternak G.W., Pintar J.E. Retention of supraspinal delta-like analgesia and loss of morphine tolerance in delta opioid receptor knockout mice. Neuron. (1999). 24: 243-252.

Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain. (1983). 16:109-10.

263