Skip to main content

Dopamine Receptors and the Treatment of Parkinson’s Disease

  • Chapter
  • First Online:

Part of the book series: The Receptors ((REC))

Abstract

Parkinson’s disease is a devastating disorder caused by progressive degeneration of dopaminergic neurons in the substantia nigra and the consequent loss of dopamine in the striatum. Dopamine replacement therapy with the dopamine precursor levodopa (l-DOPA), introduced in the 1960s, remains the most effective treatment. Unfortunately, l-DOPA, upon long-term administration, gradually loses its efficacy and eventually leads to severe motor complications, including dyskinesia. The data from numerous studies on Parkinson’s patients and animal models of the disease show a complex pattern of changes in multiple signaling pathways in the striatum induced by dopamine depletion. These include modulation of the expression and activity of several subtypes of dopamine receptors, G proteins, effectors, multiple protein kinases, components of the machinery for desensitization and trafficking of G protein-coupled receptors, ionotropic glutamate receptors, and transcription factors. Dopamine replacement therapy reverses many of these changes. However, select signaling effects are exacerbated and/or induced de novo by chronic treatment with l-DOPA. The l-DOPA-induced dyskinesia appears closely associated with selective increases in the activity of specific D1 receptor-dependent pathways. The contribution of D2 and D3 receptor-mediated signaling to dyskinesia development remains largely unexplored. The mechanisms underlying the further enhancement by l-DOPA of signaling pathways already made supersensitive by dopamine depletion need to be elucidated. The recently introduced long-lived dopamine agonists cause less dyskinesia than l-DOPA but are also less efficacious as antiparkinsonian agents. The clinically used DA agonists, which target D2-like receptors and often show preference for the D3 over D2 subtype, in addition to their antiparkinsonian action, may protect surviving dopaminergic neurons. Continuous delivery of l-DOPA or dopamine agonists, which mimics the physiological tonic stimulation of dopamine receptors, holds the promise of providing therapeutic benefits with minimal side effects. A much better understanding of the molecular processes underlying the therapeutic action of dopaminergic drugs and the development of dyskinesia is necessary in order to modify existing treatments and/or devise new approaches to maximize the beneficial effects of dopamine replacement and minimize the side effects.

This is a preview of subscription content, log in via an institution.

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   229.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   299.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   299.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

References

  1. Albin RL, Young AB, Penney JB. The functional anatomy of basal ganglia disorders. Trends Neurosci 1989;12:366–75.

    PubMed  CAS  Google Scholar 

  2. Bergman H, Deuschl G. Pathophysiology of Parkinson’s disease: from clinical neurology to basic neuroscience and back. Mov Disord 2002;17:S28–S40.

    Google Scholar 

  3. Wilms H, Sievers J, Deuschl G. Animal models of tremor. Mov Disord 1999;14:557–71.

    PubMed  CAS  Google Scholar 

  4. Aubert I, Ghorayeb I, Normand E, Bloch B. Phenotypical characterization of the neurons expressing the D1 and D2 dopamine receptors in the monkey striatum. J Comp Neurol 2000;418:22–32.

    PubMed  CAS  Google Scholar 

  5. Gerfen CR. Dopamine-mediated gene regulation in models of Parkinson’s disease. Ann Neurol 2000;47(Suppl 1):S42–S50.

    Google Scholar 

  6. Yung KK, Bolam JP, Smith AD, Hersch SM, Ciliax BJ, Levey AI. Immunocytochemical localization of D1 and D2 dopamine receptors in the basal ganglia of the rat: light and electron microscopy. Neuroscience 1995;65:709–30.

    PubMed  CAS  Google Scholar 

  7. Hurd YL, Suzuki M, Sedvall GC. D1 and D2 dopamine receptor mRNA expression in whole hemisphere sections of the human brain. J Chem Neuroanat 2001;22:127–37.

    PubMed  CAS  Google Scholar 

  8. Meador-Woodruff JH, Damask SP, Wang J, Haroutunian V, Davis KL, Watson SJ. Dopamine receptor mRNA expression in human striatum and neocortex. Neuropsychopharmacology 1996;15:17.

    PubMed  CAS  Google Scholar 

  9. Le Moine C, Bloch B. D1 and D2 dopamine receptor gene expression in the rat striatum: sensitive cRNA probes demonstrate prominent segregation of D1 and D2 mRNAs in distinct neuronal populations of the dorsal and ventral striatum. J Comp Neurol 1995;355:418–26.

    PubMed  Google Scholar 

  10. Nadjar A, Brotchie JM, Guigoni C, et al. Phenotype of striatofugal medium spiny neurons in parkinsonian and dyskinetic nonhuman primates: A call for a reappraisal of the functional organization of the basal ganglia. J Neurosci 2006;26:8653–61.

    PubMed  CAS  Google Scholar 

  11. Gurevich EV, Joyce JN. Distribution of dopamine D3 receptor expressing neurons in the human forebrain: comparison with D2 receptor expressing neurons. Neuropsy-chopharmacology 1999;20:60–80.

    PubMed  CAS  Google Scholar 

  12. Gurevich EV, Himes JW, Joyce JN. Developmental regulation of expression of the D3 dopamine receptor in rat nucleus accumbens and islands of Calleja. J Pharmacol Exp Ther 1999;289:587–98.

    PubMed  CAS  Google Scholar 

  13. Sesack SR, Aoki C, Pickel VM. Ultrastructural localization of D2 receptor-like immunoreactivity in midbrain dopamine neurons and their striatal targets. J Neurosci 1994;14:88–106.

    PubMed  CAS  Google Scholar 

  14. Levey AI, Hersch SM, Rye DB, et al. Localization of D1 and D2 dopamine receptors in brain with subtype-specific antibodies. Proc Natl Acad Sci USA 1993;90:8861–5.

    PubMed  CAS  Google Scholar 

  15. Wang H, Pickel VM. Dopamine D2 receptors are present in prefrontal cortical afferents and their targets in patches of the rat caudate-putamen nucleus. J Comp Neurol 2002;442: 392–404.

    PubMed  CAS  Google Scholar 

  16. Cepeda C, Hurst RS, Altemus KL, et al. Facilitated glutamatergic transmission in the striatum of D2 dopamine receptor-deficient mice. J Neurophysiol 2001;85:659–70.

    PubMed  CAS  Google Scholar 

  17. Gerfen CR, Keefe KA, Gauda EB. D1 and D2 dopamine receptor function in the striatum: Coactivation of D1- and D2-dopamine receptors on separate populations of neurons results in potentiated immediate early gene response in D1-containing neurons. J Neurosci 1995;15:8167–76.

    PubMed  CAS  Google Scholar 

  18. Gerfen CR, Miyachi S, Paletzki R, Brown P. D1 dopamine receptor supersensitivity in the dopamine-depleted striatum results from a switch in the regulation of ERK1/2 kinase. J Neurosci 2002;22:5042–54.

    PubMed  CAS  Google Scholar 

  19. Aizman O, Brismar H, Uhlen P, et al. Anatomical and physiological evidence for D1 and D2 dopamine receptor colocalization in neostriatal neurons. Nat Neurosci 2000;3:226.

    PubMed  CAS  Google Scholar 

  20. Surmeier DJ, Song WJ, Yan Z. Coordinated expression of dopamine receptors in neostriatal medium spiny neurons. J Neurosci 1996;16:6579–91.

    PubMed  CAS  Google Scholar 

  21. Levesque M, Parent A. The striatofugal fiber system in primates: A reevaluation of its organization based on single-axon tracing studies. Proc Natl Acad Sci USA 2005;102:11888–93.

    PubMed  CAS  Google Scholar 

  22. Wu Y, Richard S, Parent A. The organization of the striatal output system: a single-cell juxtacellular labeling study in the rat. Neurosci Res 2000;38:49–62.

    PubMed  CAS  Google Scholar 

  23. Mizuno T, Schmauss C, Rayport S. Distinct roles of presynaptic dopamine receptors in the differential modulation of the intrinsic synapses of medium-spiny neurons in the nucleus accumbens. BMC Neurosci 2007; (in press).

    Google Scholar 

  24. Bergson C, Mrzljak L, Smiley JF, Pappy M, Levenson R, Goldman-Rakic PS. Regional, cellular, and subcellular variations in the distribution of D1 and D5 dopamine receptors in primate brain. J Neurosci 1995;15:7821–36.

    PubMed  CAS  Google Scholar 

  25. Yan Z, Song W-J, Surmeier DJ. D2 dopamine receptors reduce N-type Ca2+ currents in rat neostriatal cholinergic interneurons through a membrane-delimited, protein-kinase-C-insensitive pathway. J Neurophysiol 1997;77:1003–15.

    PubMed  CAS  Google Scholar 

  26. Ding J, Guzman JN, Tkatch T, et al. RGS4-dependent attenuation of M4 autoreceptor function in striatal cholinergic interneurons following dopamine depletion. Nat Neurosci 2006;9:832–42.

    PubMed  CAS  Google Scholar 

  27. Meador-Woodruff JH, Grandy DK, Van Tol HH, et al. Dopamine receptor gene expression in the human medial temporal lobe. Neuropsychopharmacology 1994;10:239–48.

    PubMed  CAS  Google Scholar 

  28. Bouthenet ML, Souil E, Martres MP, Sokoloff P, Giros B, Schwartz JC. Localization of dopamine D3 receptor mRNA in the rat brain using in situ hybridization histochemistry: comparison with dopamine D2 receptor mRNA. Brain Res 1991;564:203–19.

    PubMed  CAS  Google Scholar 

  29. Sokoloff P, Giros B, Martres MP, Bouthenet ML, Schwartz JC. Molecular cloning and characterization of a novel dopamine receptor (D3) as a target for neuroleptics. Nature 1990;347:146–51.

    PubMed  CAS  Google Scholar 

  30. Bezard E, Ferry S, Mach U, et al. Attenuation of levodopa-induced dyskinesia by normalizing dopamine D3 receptor function. Nature Med 2003;9:762–7.

    PubMed  CAS  Google Scholar 

  31. Murray AM, Ryoo HL, Gurevich E, Joyce JN. Localization of dopamine D3 receptors to mesolimbic and D2 receptors to mesostriatal regions of human forebrain. Proc Natl Acad Sci USA 1994;91:11271–5.

    PubMed  CAS  Google Scholar 

  32. Suzuki M, Hurd YL, Sokoloff P, Schwartz J-C, Sedvall G. D3 dopamine receptor mRNA is widely expressed in the human brain. Brain Res 1998;779:58–74.

    PubMed  CAS  Google Scholar 

  33. Le Moine C, Bloch B. Expression of the D3 dopamine receptor in peptidergic neurons of the nucleus accumbens: comparison with the D1 and D2 dopamine receptors. Neuroscience 1996;73:131–43.

    PubMed  Google Scholar 

  34. Perachon S, Schwartz JC, Sokoloff P. Functional potencies of new antiparkinsonian drugs at recombinant human dopamine D1, D2 and D3 receptors. Eur J Pharmacol 1999;366: 293–300.

    PubMed  CAS  Google Scholar 

  35. Gurevich EV, Bordelon Y, Shapiro RM, Arnold SE, Gur RE, Joyce JN. Mesolimbic dopamine D3 receptors and use of antipsychotics in patients with schizophrenia. A postmortem study. Arch Gen Psychiatry 1997;54:225–32.

    PubMed  CAS  Google Scholar 

  36. Diaz J, Ridray S, Mignon V, Griffon N, Schwartz JC, Sokoloff P. Selective expression of dopamine D3 receptor mRNA in proliferative zones during embryonic development of the rat brain. J Neurosci 1997;17:4282–92.

    PubMed  CAS  Google Scholar 

  37. Araki KY, Sims JR., Bhide PG. Dopamine receptor mRNA and protein expression in the mouse corpus striatum and cerebral cortex during pre- and postnatal development. Brain Res 2007;1156:31–45.

    PubMed  CAS  Google Scholar 

  38. Gurevich EV, Joyce JN. Dopamine D(3) receptor is selectively and transiently expressed in the developing whisker barrel cortex of the rat. J Comp Neurol 2000;420:35–51.

    PubMed  CAS  Google Scholar 

  39. Gurevich EV, Robertson RT, Joyce JN. Thalamo-cortical afferents control transient expression of the dopamine D(3) receptor in the rat somatosensory cortex. Cereb Cortex 2001;11:691–701.

    PubMed  CAS  Google Scholar 

  40. Rivera A, Trías S, Peñafiel A, et al. Expression of D4 dopamine receptors in striatonigral and striatopallidal neurons in the rat striatum. Brain Res 2003;989:35–41.

    PubMed  CAS  Google Scholar 

  41. Rivera A, Cuéllar B, Girón FJ, Grandy DK, de la Calle A, Moratalla R. Dopamine D4 receptors are heterogeneously distributed in the striosomes/matrix compartments of the striatum. J Neurochem 2002;80:219–29.

    PubMed  CAS  Google Scholar 

  42. Ciliax BJ, Nash N, Heilman C, et al. Dopamine D5 receptor immunolocalization in rat and monkey brain. Synapse 2000;37:125–45.

    PubMed  CAS  Google Scholar 

  43. Centonze D, Grande C, Usiello A, et al. Receptor subtypes involved in the presynaptic and postsynaptic actions of dopamine on striatal interneurons. J Neurosci 2003;23:6545–54.

    Google Scholar 

  44. Khan ZU, Gutiérrez A, Martín R, Peñafiel A, Rivera A, de la Calle A. Dopamine D5 receptors of rat and human brain. Neuroscience 2000;100:689–99.

    PubMed  CAS  Google Scholar 

  45. Centonze D, Grande C, Saulle E, et al. Distinct roles of D1 and D5 dopamine receptors in motor activity and striatal synaptic plasticity. J Neurosci 2003;23:8506–12.

    PubMed  CAS  Google Scholar 

  46. Rinne JO, Laihinen A, NÂgren K, et al. PET demonstrates different behaviour of striatal dopamine D-1 and D-2 receptors in early Parkinson’s disease. J Neurosci Res 1990;27: 494–9.

    PubMed  CAS  Google Scholar 

  47. Rinne UK, Laihinen A, Rinne JO, NaÂgren K, Bergman J, Ruotsalainen U. Positron emission tomography demonstrates dopamine D2 receptor supersensitivity in the striatum of patients with early Parkinson’s disease. Mov Disord 1990;5:55–9.

    PubMed  CAS  Google Scholar 

  48. Brooks DJ, Ibanez V, Sawle GV, et al. Striatal D2 receptor status in patients with Parkinson’s disease, striatonigral degeneration, and progressive supranuclear palsy, measured with [11C]-raclopride and positron emission tomography. Ann Neurol 1992;31:184–92.

    PubMed  CAS  Google Scholar 

  49. Laulumaa V, Kuikka JT, Soininen H, Bergstrom K, Lansimies E, Riekkinen P. Imaging of D2 dopamine receptors of patients with Parkinson’s disease using single photon emission computed tomography and iodobenzamide I 123. Arch Neurol 1993;50:509–12.

    PubMed  CAS  Google Scholar 

  50. SawleGV, Playford ED, Brooks DJ, Quinn N, Frackowiak RSJ. Asymmetrical pre-synaptic and post-synaptic changes in the striatal dopamine projection in dopa naive Parkinsonism. Diagnostic implications of the D2 receptor status. Brain 1993;116:853–67.

    PubMed  Google Scholar 

  51. Antonini A, Schwarz J, Oertel WH, Pogarell O, Leenders KL. Long-term changes of striatal dopamine D2 Receptors in patients with Parkinson’s disease: A study with positron emission tomography and [11C]Raclopride. Mov Disord 1997;12:33–8.

    PubMed  CAS  Google Scholar 

  52. Turjanski N, Lees AJ, Brooks DJ. In vivo studies on striatal dopamine D1 and D2 site binding in l- dopa-treated Parkinson’s disease patients with and without dyskinesias. Neurology 1997;49:717–23.

    PubMed  CAS  Google Scholar 

  53. Kim YJ, Ichise M, Ballinger JR, et al. Combination of dopamine transporter and D2 receptor SPECT in the diagnostic evaluation of PD, MSA, and PSP. Mov Disord 2002;17:303–12.

    PubMed  Google Scholar 

  54. Thobois S, Vingerhoets F, Fraix V, et al. Role of dopaminergic treatment in dopamine receptor down-regulation in advanced Parkinson disease: A positron emission tomographic study. Arch Neurol 2004;61:1705–9.

    PubMed  Google Scholar 

  55. Bokobza B, Ruberg M, Scatton B, Javoy-Agid F, Agid Y. [3H]spiperone binding, dopamine and HVA concentrations in Parkinson’s disease and supranuclear palsy. Eur J Pharmacol 1984;99:167.

    PubMed  CAS  Google Scholar 

  56. Joyce JN. Differential response of striatal dopamine and muscarinic cholinergic receptor subtypes to the loss of dopamine. III. Results in Parkinson’s disease cases. Brain Res 1993;600:156.

    PubMed  CAS  Google Scholar 

  57. Piggott MA, Marshall EF, Thomas N, et al. Striatal dopaminergic markers in dementia with Lewy bodies, Alzheimer’s and Parkinson’s diseases: rostrocaudal distribution. Brain 1999;122:1449–68.

    PubMed  Google Scholar 

  58. Ryoo HL, Pierrotti D, Joyce JN. Dopamine D3 receptor is decreased and D2 receptor is elevated in the striatum of Parkinson’s disease. Mov Disord 1998;13:788–97.

    PubMed  CAS  Google Scholar 

  59. Goulet M, Morissette M, Calon F, et al. Continuous or pulsatile chronic D2 dopamine receptor agonist (U91356A) treatment of drug-naive 4-phenyl-1,2,3,6-tetrahydropyridine monkeys differentially regulates brain D1 and D2 receptor expression: in situ hybridization histochemical analysis. Neuroscience 1997;79(2):497.

    PubMed  CAS  Google Scholar 

  60. Bychkov E, Ahmed MR., Dalby KN, Gurevich EV. Dopamine depletion and subsequent treatment with l-DOPA, but not the long-lived dopamine agonist pergolide, enhances activity of the Akt pathway in the rat striatum. J Neurochem 2007;102:699–711.

    PubMed  CAS  Google Scholar 

  61. Herrero MT, Augood SJ, Asensi H, et al. Effects of -DOPA-therapy on dopamine D2 receptor mRNA expression in the striatum of MPTP-intoxicated parkinsonian monkeys. Mol Brain Res 1996;42:149.

    PubMed  CAS  Google Scholar 

  62. Morissette M, Goulet M, Calon F, et al. Changes of D1 and D2 dopamine receptor mRNA in the brains of monkeys lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine: Correction with chronic administration of l-3,4-dihydroxyphenylalanine. Mol Pharmacol 1996;50:1073–9.

    PubMed  CAS  Google Scholar 

  63. Bezard E, Dovero S, Prunier C, et al. Relationship between the appearance of symptoms and the level of nigrostriatal degeneration in a progressive 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson’s disease. J Neurosci 2001;21:6853–61.

    PubMed  CAS  Google Scholar 

  64. Shinotoh H, Inoue O, Hirayama K, et al. Dopamine D1 receptors in Parkinson’s disease and striatonigral degeneration: A positron emission tomography study. J Neurol Neurosurg Psychiat 1993;56:467–72.

    PubMed  CAS  Google Scholar 

  65. Lévesque D, Martres M-P, Diaz J, et al. A paradoxical regulation of the dopamine D3 receptor expression suggests the involvement of an anterograde factor from dopamine neurons. Proc Nat Acad Sci USA 1995;92:1719–23.

    PubMed  Google Scholar 

  66. Bordet R, Ridray S, Carboni C, Diaz J, Sokoloff P, Schwartz JC. Induction of dopamine D3 receptor expression as a mechanism of behavioral sensitization to levodopa. Proc Natl Acad Sci USA 1997;94:3363–7.

    PubMed  CAS  Google Scholar 

  67. Bordet R, Ridray S, Schwartz JC, Sokoloff P. Involvement of the direct striatonigral pathway in levodopa-induced sensitization in 6-hydroxydopamine-lesioned rats. Eur J Neurosci 2000;12:2117–23.

    PubMed  CAS  Google Scholar 

  68. Quik M, Police S, He L, Di Monte DA, Langston JW. Expression of D(3) receptor messenger RNA and binding sites in monkey striatum and substantia nigra after nigrostriatal degeneration: effect of levodopa treatment. Neuroscience 2000;98:263–73.

    PubMed  CAS  Google Scholar 

  69. Hirsch E, Graybiel AM, Agid YA. Melanized dopaminergic neurons are differentially susceptible to degeneration in Parkinson’s disease. Nature 1988;334:345–8.

    PubMed  CAS  Google Scholar 

  70. Goto S, Hirano A, Matsumoto S. Subdivisional involvement of nigrostriatal loop in idiopathic Parkinson’s disease and striatonigral degeneration. Ann Neurol 1989;26: 766–70.

    PubMed  CAS  Google Scholar 

  71. Gibb WR, Lees AJ. Anatomy, pigmentation, ventral and dorsal subpopulations of the substantia nigra, and differential cell death in Parkinson’s disease. J Neurol Neurosurg Psychiatry 1991;54:388–96.

    PubMed  CAS  Google Scholar 

  72. Bychkov E, Gurevich VV, Joyce JN, Benovic JL, Gurevich EV. Arrestins and two receptor kinases are upregulated in Parkinson’s disease with dementia. Neurobiol Aging 2008;29:379–96.

    PubMed  CAS  Google Scholar 

  73. Joyce JN, Ryoo HL, Beach TB, et al. Loss of response to levodopa in Parkinson’s disease and co-occurrence with dementia: role of D3 and not D2 receptors. Brain Res 2002;955: 138–52.

    PubMed  CAS  Google Scholar 

  74. Kaasinen V, Aalto S, Någren K, Hietala J, Sonninen P, Rinne JO. Extrastriatal dopamine D2 receptors in Parkinson’s disease: a longitudinal study. J Neural Transm 2003;110:591–601.

    PubMed  CAS  Google Scholar 

  75. Ungerstedt U. Postsynaptic supersensitivity after 6-hydroxydopamine induced degeneration of the nigrostriatal dopamine system. Acta Physiol Scand Suppl 1971;367:69–93.

    PubMed  CAS  Google Scholar 

  76. Kim DS, Szczypka MS, Palmiter RD. Dopamine-deficient mice are hypersensitive to dopamine receptor agonists. J Neurosci 2000;20:4405–13.

    PubMed  CAS  Google Scholar 

  77. Cai G, Wang HY, Friedman E. Increased dopamine receptor signaling and dopamine receptor-G protein coupling in denervated striatum. J Pharmacol Exp Ther 2002;302: 1105–12.

    PubMed  CAS  Google Scholar 

  78. Corvol J-C, Muriel M-P, Valjent E, et al. Persistent increase in olfactory type G-protein alpha subunit levels may underlie D1 receptor functional hypersensitivity in Parkinson disease. J Neurosci 2004;24:7007–14.

    PubMed  CAS  Google Scholar 

  79. Mishra RK, Gardner EL, Katzman R, Makman MH. Enhancement of dopamine-stimulated adenylate cyclase activity in rat caudate after lesions in substantia nigra: evidence for denervation supersensitivity. Proc Natl Acad Sci USA 1974;71:3883–7.

    PubMed  CAS  Google Scholar 

  80. Mishra RK, Marshall AM, Varmuza SL. Supersensitivity in rat caudate nucleus: effects of 6-hydroxydopamine on the time course of dopamine receptor and cyclic AMP changes. Brain Res 1980;200:47–57.

    PubMed  CAS  Google Scholar 

  81. Pifl C, Nanoff C, Schingnitz G, Schütz W, Hornykiewicz O. Sensitization of dopamine-stimulated adenylyl cyclase in the striatum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated rhesus monkeys and patients with idiopathic Parkinson’s disease. J Neurochem 1992;58:1997–2004.

    PubMed  CAS  Google Scholar 

  82. Pifl C, Reither H, Hornykiewicz O. Functional sensitization of striatal dopamine D1 receptors in the 6-hydroxydopamine-lesioned rat. Brain Res 1992;572:87–93.

    PubMed  CAS  Google Scholar 

  83. Satoh H, Satoh Y, Notsu Y, Honda F. Adenosine 3',5'-cyclic monophosphate as a possible mediator of rotational behaviour induced by dopaminergic receptor stimulation in rats lesioned unilaterally in the substantia nigra. Eur J Pharmacol 1976;39:365–77.

    PubMed  CAS  Google Scholar 

  84. Tong J, Fitzmaurice PS, Ang LC, Furukawa Y, Guttman M, Kish SJ. Brain dopamine-stimulated adenylyl cyclase activity in Parkinson’s disease, multiple system atrophy, and progressive supranuclear palsy. Ann Neurol 2004;55:125–9.

    PubMed  CAS  Google Scholar 

  85. Santini E, Valjent E, Usiello A, et al. Critical involvement of cAMP/DARPP-32 and extracellular signal-regulated protein kinase signaling in l-DOPA-induced dyskinesia. J Neurosci 2007;27:6995–7005.

    PubMed  CAS  Google Scholar 

  86. Ouimet CC, Greengard P. Distribution of DARPP-32 in the basal ganglia: an electron microscopic study. J Neurocytol 1990;19:39–52.

    PubMed  CAS  Google Scholar 

  87. Hemmings HCJ, Williams RR, Konigsberg WH, Greengard P. DARPP-32, a dopamine- and adenosine 3':5'-monophosphate-regulated neuronal phosphoprotein. I. Amino acid sequence around the phosphorylated threonine. J Biol Chem 1984;259:14486–90.

    PubMed  CAS  Google Scholar 

  88. Fienberg AA, Hiroi N, Mermelstein PG, et al. DARPP-32: Regulator of the efficacy of dopaminergic neurotransmission. Science 1998;281:838–42.

    PubMed  CAS  Google Scholar 

  89. Bibb JA, Snyder GL, Nishi A, et al. Phosphorylation of DARPP-32 by Cdk5 modulates dopamine signalling in neurons. Nature 1999;402:669.

    PubMed  CAS  Google Scholar 

  90. Nishi A, Bibb JA, Snyder GL, Higashi H, Nairn AC, Greengard P. Amplification of dopaminergic signaling by a positive feedback loop. Proc Natl Acad Sci USA 2000;97:12840–5.

    PubMed  CAS  Google Scholar 

  91. Brown A, Deutch AY, Colbran RJ. Dopamine depletion alters phosphorylation of striatal proteins in a model of Parkinsonism. Eur J Neurosci 2005;22:247–56.

    PubMed  Google Scholar 

  92. Svenningsson P, Lindskog M, Ledent C, et al. Regulation of the phosphorylation of the dopamine- and cAMP-regulated phosphoprotein of 32 kDa in vivo by dopamine D1, dopamine D2, and adenosine A2A receptors. Proc Natl Acad Sci USA 2000;97:1856–60.

    PubMed  CAS  Google Scholar 

  93. Westin JE, Vercammen L, Strome EM, Konradi C, Cenci MA. Spatiotemporal pattern of striatal ERK1/2 phosphorylation in a rat model of l-DOPA-induced dyskinesia and the role of dopamine D1 receptors. Biol Psychiatry 2007;62:800–10.

    PubMed  CAS  Google Scholar 

  94. Emborg-Knott ME, Domino EF. MPTP-induced hemiparkinsonism in nonhuman primates 6–8 years after a single unilateral intracarotid dose. Exp Neurol 1998;152:214–20.

    PubMed  CAS  Google Scholar 

  95. Aubert I, Guigoni C, Hakansson K, et al. Increased D1 dopamine receptor signaling in levodopa-induced dyskinesia. Ann Neurol 2005;57:17–26.

    PubMed  CAS  Google Scholar 

  96. Bezard E, Gross CE, Qin L, Gurevich VV, Benovic JL, Gurevich EV. l-DOPA reverses the MPTP-induced elevation of the arrestin2 and GRK6 expression and enhanced ERK activation in monkey brain. Neurobiol Dis 2005;18:323–35.

    PubMed  CAS  Google Scholar 

  97. Svenningsson P, Arts J, Gunne L, Andren PE. Acute and repeated treatment with -DOPA increase c-jun expression in the 6-hydroxydopamine-lesioned forebrain of rats and common marmosets. Brain Res 2002;955:8–15.

    PubMed  CAS  Google Scholar 

  98. Hess J, Angel P, Schorpp-Kistner M.AP-1 subunits: quarrel and harmony among siblings. J Cell Sci 2004;117:5965–73.

    PubMed  CAS  Google Scholar 

  99. Chinenov Y, Kerppola TK. Close encounters of many kinds: Fos-Jun interactions that mediate transcription regulatory specificity. Oncogene 2001;20:2438–52.

    PubMed  CAS  Google Scholar 

  100. Berke JD, Paletzki R, Aronson GJ, Hyman SE, Gerfen CR. A complex program of striatal gene expression induced by dopaminergic stimulation. J Neurosci 1998;18:5301–10.

    PubMed  CAS  Google Scholar 

  101. Cenci MA, Tranberg A, Andersson M, Hilbertson A. Changes in the regional and compartmental distribution of FosB- and JunB-like immunoreactivity induced in the dopamine-denervated rat striatum by acute or chronic l-DOPA treatment. Neuroscience 1999;94:515.

    PubMed  CAS  Google Scholar 

  102. Valastro B, Andersson M, Lindgren HS, Cenci MA. Expression pattern of JunD after acute or chronic l-DOPA treatment: Comparison with DeltaFosB. Neuroscience 2007;144:198–207.

    PubMed  CAS  Google Scholar 

  103. Shaywitz AJ, Greenberg ME. CREB: A stimulus-induced transcription factor activated by a diverse array of extracellular signals. Ann Rev Biochem 1999;68:821–61.

    PubMed  CAS  Google Scholar 

  104. Mayr B, Montminy M. Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat Rev Mol Cell Biol 2001;2:599–609.

    PubMed  CAS  Google Scholar 

  105. OhJD, Chartisathian K, Ahmed SM, Chase TN. Cyclic AMP responsive element binding protein phosphorylation and persistent expression of levodopa-induced response alterations in unilateral nigrostriatal 6-OHDA lesioned rats. J Neurosci Res 2003;72:768–80.

    PubMed  CAS  Google Scholar 

  106. Cole DG, Kobierski LA, Konradi C, Hyman SE. 6-Hydroxydopamine lesions of rat substantia nigra up-regulate dopamine-induced phosphorylation of the cAMP-response element-binding protein in striatal neurons. Proc Natl Acad Sci USA 1994;91:9631–5.

    PubMed  CAS  Google Scholar 

  107. Konradi C, Cole RL, Heckers S, Hyman SE. Amphetamine regulates gene expression in rat striatum via transcription factor CREB. J Neurosci 1994;14:5623–34.

    PubMed  CAS  Google Scholar 

  108. Andersson M, Konradi C, Cenci MA. cAMP response element-binding protein is required for dopamine-dependent gene expression in the intact but not the dopamine-denervated striatum. J Neurosci 2001;21:9930–43.

    PubMed  CAS  Google Scholar 

  109. Picconi B, Gardoni F, Centonze D, et al. Abnormal Ca2+-calmodulin-dependent protein kinase II function mediates synaptic and motor deficits in experimental parkinsonism. J Neurosci 2004;24:5283–91.

    PubMed  CAS  Google Scholar 

  110. Cull-Candy S, Brickley S, Farrant M. NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol 2001;11:327.

    PubMed  CAS  Google Scholar 

  111. Chen B-S, Roche KW. Regulation of NMDA receptors by phosphorylation. Neuropharmacology 2007;53:362.

    PubMed  Google Scholar 

  112. Dunah AW, Wang Y, Yasuda RP, et al. Alterations in subunit expression, composition, and phosphorylation of striatal N-methyl-D-aspartate glutamate receptors in a rat 6-hydroxydopamine model of Parkinson’s disease. Mol Pharmacol 2000;57:342–52.

    PubMed  CAS  Google Scholar 

  113. Hallett PJ, Dunah AW, Ravenscroft P, et al. Alterations of striatal NMDA receptor subunits associated with the development of dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Neuropharmacology 2005;48:503–16.

    PubMed  CAS  Google Scholar 

  114. Fiorentini C, Rizzetti MC, Busi C, et al. Loss of synaptic D1 dopamine/N-methyl-D-aspartate glutamate receptor complexes in l-DOPA-induced dyskinesia in the rat. Mol Pharmacol 2006;69:805–12.

    PubMed  CAS  Google Scholar 

  115. Oh JD, Vaughan CL, Chase TN. Effect of dopamine denervation and dopamine agonist administration on serine phosphorylation of striatal NMDA receptor subunits. Brain Res 1999;821:433–42.

    PubMed  CAS  Google Scholar 

  116. Gardoni F, Picconi B, Ghiglieri V, et al. A Critical Interaction between NR2B and MAGUK in l-DOPA Induced Dyskinesia. J Neurosci 2006;26:2914–22.

    PubMed  CAS  Google Scholar 

  117. Oh JD, Russell DS, Vaughan CL, Chase TN. Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and l-DOPA administration. Brain Res 1998;813:150–9.

    PubMed  CAS  Google Scholar 

  118. Ahmed MR, Bychkov E, Gurevich VV, Benovic JL, Gurevich EV. Altered expression and subcellular distribution of GRK subtypes in the dopamine-depleted rat basal ganglia is not normalized by l-DOPA treatment. J Neurochem 2007;104:1622–36.

    PubMed  Google Scholar 

  119. Nash JE, Johnston TH, Collingridge GL, Garner CC, Brotchie JM. Subcellular redistribution of the synapse-associated proteins PSD-95 and SAP97 in animal models of Parkinson’s disease and l-DOPA-induced dyskinesia. FASEB J 2005;19:583–5.

    PubMed  CAS  Google Scholar 

  120. Picconi B, Centonze D, Hakansson K, et al. Loss of bidirectional striatal synaptic plasticity in l-DOPA-induced dyskinesia. Nat Neurosci 2003;6:501–6.

    PubMed  CAS  Google Scholar 

  121. Picconi B, Paille V, Ghiglieri V, et al. l-DOPA dosage is critically involved in dyskinesia via loss of synaptic depotentiation. Neurobiol Dis 2008;29:327.

    PubMed  CAS  Google Scholar 

  122. Wang H, Katz J, Dagostino P, Soghomonian JJ. Unilateral 6-hydroxydopamine lesion of dopamine neurons and subchronic l-DOPA administration in the adult rat alters the expression of the vesicular GABA transporter in different subsets of striatal neurons and in the substantia nigra, pars reticulata. Neuroscience 2007;145:727–37.

    PubMed  CAS  Google Scholar 

  123. Soghomonian J-J, Laprade N. Glutamate decarboxylase (GAD67 and GAD65) gene expression is increased in a subpopulation of neurons in the putamen of parkinsonian monkeys. Synapse 1997;27:122–32.

    PubMed  CAS  Google Scholar 

  124. Carta AR, Fenu S, Pala P, Tronci E, Morelli M. Selective modifications in GAD67 mRNA levels in striatonigral and striatopallidal pathways correlate to dopamine agonist priming in 6-hydroxydopamine-lesioned rats. Eur J Neurosci 2003;18:2563–72.

    PubMed  CAS  Google Scholar 

  125. Lindgren HS, Rylander D, Ohlin KE, Lundblad M, Cenci MA. The "motor complication syndrome" in rats with 6-OHDA lesions treated chronically with l-DOPA: relation to dose and route of administration. Behav Brain Res 2007;177:150–9.

    PubMed  CAS  Google Scholar 

  126. Engber TM, Marin C, Susel Z, Chase TN. Differential effects of chronic dopamine D1 and D2 receptor agonists on rotational behavior and dopamine receptor binding. Eur J Pharmacol 1993;236:385–93.

    PubMed  CAS  Google Scholar 

  127. Löschmann PA, Wüllner U, Heneka MT, et al. Differential interaction of competitive NMDA and AMPA antagonists with selective dopamine D-1 and D-2 agonists in a rat model of Parkinson’s disease. Synapse 1997;26:381–91.

    PubMed  Google Scholar 

  128. Paul ML, Graybiel AM, David JC, Robertson HA. D1-like and D2-like dopamine receptors synergistically activate rotation and c-fos expression in the dopamine-depleted striatum in a rat model of Parkinson’s disease. J Neurosci 1992;12:3729–42.

    PubMed  CAS  Google Scholar 

  129. Cai G, Zhen X, Uryu K, Friedman E. Activation of extracellular signal-regulated protein kinases is associated with a sensitized locomotor response to D(2) dopamine receptor stimulation in unilateral 6-hydroxydopamine-lesioned rats. J Neurosci 2000;20: 1849–57.

    PubMed  CAS  Google Scholar 

  130. Newman-Tancredi A, Cussac D, Brocco M, et al. Dopamine D2 receptor-mediated G-protein activation in rat striatum: functional autoradiography and influence of unilateral 6-hydroxydopamine lesions of the substantia nigra. Brain Res 2001;920:41.

    Google Scholar 

  131. LaHoste GJ, Marshall JF. Rapid development of D1 and D2 dopamine receptor supersensitivity as indicated by striatal and pallidal Fos expression. Neurosci Lett 1994;179: 153–6.

    PubMed  CAS  Google Scholar 

  132. Marshal JF, Cole BN, LaHoste GJ. Dopamine D2 receptor control of pallidal fos expression: comparisons between intact and 6-hydroxydopamine-treated hemispheres. Brain Res 1993;632:308–13.

    Google Scholar 

  133. Zhen X, Torres C, Cai G, Friedman E. Inhibition of protein tyrosine/mitogen-activated protein kinase phosphatase activity is associated with D2 dopamine receptor supersensitivity in a rat model of Parkinson’s disease. Mol Pharmacol 2002;62:1356–63.

    PubMed  CAS  Google Scholar 

  134. Bamford NS, Robinson S, Palmiter RD, Joyce JA, Moore C, Meshul CK. Dopamine modulates release from corticostriatal terminals. J Neurosci 2004;24:9541–52.

    PubMed  CAS  Google Scholar 

  135. Picconi B, Centonze D, Rossi S, Bernardi G, Calabresi P. Therapeuticdoses of l-dopa reverse hypersensitivity of corticostriatal D2-dopamine receptors and glutamatergic overactivity in experimental parkinsonism. Brain 2004;127:1661–9.

    PubMed  Google Scholar 

  136. Lindefors N. Dopaminergic regulation of glutamic acid decarboxylase mRNA expression and GABA release in the striatum: A review. Progr Neuro-Psychopharmacol Biol Psychiatry 1993;17:887.

    CAS  Google Scholar 

  137. Gerfen CR, McGinty JF, Young WS. Dopamine differentially regulates dynorphin, substance P, and enkephalin expression in striatal neurons: in situ hybridization histochemical analysis. J Neurosci 1991;11:1016–31.

    PubMed  CAS  Google Scholar 

  138. Henry B, Crossman AR, Brotchie JM. Effect of repeated l-DOPA, bromocriptine, or lisuride administration on preproenkephalin-A and preproenkephalin-B mRNA levels in the striatum of the 6-hydroxydopamine-lesioned rat. Exp Neurol 1999;155:204–20.

    PubMed  CAS  Google Scholar 

  139. Herrero MT, Augood SJ, Hirsch EC, et al. Effects of l-DOPA on preproenkephalin and preprotachykinin gene expression in the MPTP-treated monkey striatum. Neuroscience 1995;68:1189–98.

    PubMed  CAS  Google Scholar 

  140. Morissette M, Goulet M, Soghomonian JJ, et al. Preproenkephalin mRNA expression in the caudate-putamen of MPTP monkeys after chronic treatment with the D2 agonist U91356A in continuous or intermittent mode of administration: comparison with l-DOPA therapy. Brain Res Mol Brain Res 1997;49:55–62.

    PubMed  CAS  Google Scholar 

  141. Morissette M, Grondin R, Goulet M, Bédard PJ, Di Paolo T. Differential regulation of striatal preproenkephalin and preprotachykinin mRNA levels in MPTP-lesioned monkeys chronically treated with dopamine D1 or D2 agonists. J Neurochem 1999;72:682–92.

    PubMed  CAS  Google Scholar 

  142. Westin JE, Andersson M, Lundblad M, Cenci MA. Persistent changes in striatal gene expression induced by long-term l-DOPA treatment in a rat model of Parkinson’s disease. Eur J Neurosci 2001;14:1171–6.

    PubMed  CAS  Google Scholar 

  143. St-Hilaire M, Bourhis E, Lévesque D, Rouillard C. Impaired behavioural and molecular adaptations to dopamine denervation and repeated l-DOPA treatment in Nur77-knockout mice. Eur J Neurosci 2006;24:795–805.

    PubMed  Google Scholar 

  144. St-Hilaire M, Landry E, Levesque D, Rouillard C. Denervation and repeated l-DOPA induce a coordinate expression of the transcription factor NGFI-B in striatal projection pathways in hemi-parkinsonian rats. Neurobiol Dis 2003;14:98–109.

    PubMed  CAS  Google Scholar 

  145. St-Hilaire M, Landry E, Levesque D, Rouillard C. Denervation and repeated l-DOPA induce complex regulatory changes in neurochemical phenotypes of striatal neurons: Implication of a dopamine D1-dependent mechanism. Neurobiol Dis 2005;20:450.

    PubMed  CAS  Google Scholar 

  146. Ethier I, Beaudry G, St-Hilaire M, Milbrandt J, Rouillard C, Lévesque D. The transcription factor NGFI-B (Nur77) and retinoids play a critical role in acute neuroleptic-induced extrapyramidal effect and striatal neuropeptide gene expression. Neuropsychopharmacology 2004;29:335–46.

    PubMed  CAS  Google Scholar 

  147. Scholz B, Svensson M, Alm H, et al. Striatal proteomic analysis suggests that first l-Dopa dose equates to chronic exposure. PLoS ONE 2008;3:e1589.

    PubMed  Google Scholar 

  148. Pollack AE, Yates TM. Prior D1 dopamine receptor stimulation is required to prime D2-mediated striatal Fos expression in 6-hydroxydopamine-lesioned rats. Neuroscience 1999;94:505–14.

    PubMed  CAS  Google Scholar 

  149. Pollack AE, Turgeon SM, Fink JS. Apomorphine priming alters the response of striatal outflow pathways to D2 agonist stimulation in 6-hydroxydopamine-lesioned rats. Neuroscience 1997;79:79.

    PubMed  CAS  Google Scholar 

  150. Pollack AE, Strauss JB. Time dependence and role of N-methyl-D-aspartate glutamate receptors in the priming of D2-mediated rotational behavior and striatal Fos expression in 6-hydroxydopamine lesioned rats. Brain Res 1999;827:160–8.

    PubMed  CAS  Google Scholar 

  151. Paul ML, Currie RW, Robertson HA. Priming of a D1 dopamine receptor behavioural response is dissociated from striatal immediate-early gene activity. Neuroscience 1995;66:347–59.

    PubMed  CAS  Google Scholar 

  152. Wang Z, Kai L, Day M, et al. Dopaminergic control of corticostriatal long-term synaptic depression in medium spiny neurons is mediated by cholinergic interneurons. Neuron 2006;50:443–52.

    PubMed  CAS  Google Scholar 

  153. Herve D, Levi-Strauss M, Marey-Semper I, et al. Golf and Gs in rat basal ganglia: possible involvement of Golf in the coupling of dopamine D1 receptor with adenylyl cyclase. J Neurosci 1993;13:2237–48.

    PubMed  CAS  Google Scholar 

  154. Corvol JC, Studler JM, Schonn JS, Girault JA, Herve D. Gaolf is necessary for coupling D1 and A2a receptors to adenylyl cyclase in the striatum. J Neurochem 2001;76:1585–8.

    PubMed  CAS  Google Scholar 

  155. Herve D, Le Moine C, Corvol J-C, et al. Gaolf levels are regulated by receptor usage and control dopamine and adenosine action in the striatum. J Neurosci 2001;21:4390–9.

    PubMed  CAS  Google Scholar 

  156. Nishino N, Kitamura N, Hashimoto T, Tanaka C. Transmembrane signalling systems in the brain of patients with Parkinson’s disease. Rev Neurosci 1993;4:213–22.

    PubMed  CAS  Google Scholar 

  157. Gurevich VV, Gurevich EV. The structural basis of arrestin-mediated regulation of G-protein-coupled receptors. Pharmacology & Therapeutics 2006;110:465–502.

    CAS  Google Scholar 

  158. Krasel C, Vilardaga JP, Bunemann M, Lohse MJ. Kinetics of G-protein-coupled receptor signalling and desensitization. Biochem Soc Trans 2004;32:1029–31.

    PubMed  CAS  Google Scholar 

  159. Violin JD, DiPilato LM, Yildirim N, Elston TC, Zhang J, Lefkowitz RJ. β2-Adrenergic receptor signaling and desensitization elucidated by quantitative modeling of real time cAMP dynamics. J Biol Chem 2008;283:2949–61.

    PubMed  CAS  Google Scholar 

  160. Bohn LM, Gainetdinov RR, Sotnikova TD, et al. Enhanced rewarding properties of morphine, but not cocaine, in beta-arrestin-2 knock-out mice. J Neurosci 2003;23(32):10265–73.

    PubMed  CAS  Google Scholar 

  161. Bohn LM, Lefkowitz RJ, Caron MG. Differential mechanisms of morphine antinociceptive tolerance revealed in beta-arrestin-2 knock-out mice. J Neurosci 2002;22:10494–500.

    PubMed  CAS  Google Scholar 

  162. Bohn LM, Lefkowitz RJ, Gainetdinov RR, Peppel K, Caron MG, Lin FT. Enhanced morphine analgesia in mice lacking beta-arrestin2. Science 1999;286:2495–8.

    PubMed  CAS  Google Scholar 

  163. Gainetdinov RR, Bohn LM, Sotnikova TD, et al. Dopaminergic supersensitivity in G protein-coupled receptor kinase 6-deficient mice. Neuron 2003;38:291–303.

    PubMed  CAS  Google Scholar 

  164. Gainetdinov RR, Bohn LM, Walker JK, et al. Muscarinic supersensitivity and impaired receptor desensitization in G protein-coupled receptor kinase 5-deficient mice. Neuron 1999;24:1029–36.

    PubMed  CAS  Google Scholar 

  165. Pan L, Gurevich EV, Gurevich VV. The nature of the arrestin x receptor complex determines the ultimate fate of the internalized receptor. J Biol Chem 2003;278:11623–32.

    PubMed  CAS  Google Scholar 

  166. Willets JM, Nash MS, Challiss RA, Nahorski SR. Imaging of muscarinic acetylcholine receptor signaling in hippocampal neurons: evidence for phosphorylation-dependent and -independent regulation by G-protein-coupled receptor kinases. J Neurosci 2004;24:4157–62.

    PubMed  CAS  Google Scholar 

  167. Willets JM, Parent JL, Benovic JL, Kelly E. Selective reduction in A2 adenosine receptor desensitization following antisense-induced suppression of G protein-coupled receptor kinase 2 expression. J Neurochem 1999;73:1781–9.

    PubMed  CAS  Google Scholar 

  168. Berman DM, Gilman AG. Mammalian RGS proteins: Barbarians at the gate. J Biol Chem 1998;273:1269–72.

    PubMed  CAS  Google Scholar 

  169. Ross EM, Wilkie TM. GTPase-activating proteins for heterotrimeric G proteins: regulators of G protein signaling (RGS) and RGS-like proteins. Annu Rev Biochem 2000;69:795–827.

    PubMed  CAS  Google Scholar 

  170. Gold SJ, Ni YG, Dohlman HG, Nestler EJ. Regulators of G-protein signaling (RGS) proteins: Region-specific expression of nine subtypes in rat brain. J Neurosci 1997;17: 8024–37.

    PubMed  CAS  Google Scholar 

  171. Granneman JG, Zhai Y, Zhu Z, et al. Molecular characterization of human and rat RGS 9L, a novel splice variant enriched in dopamine target regions, and chromosomal localization of the RGS 9 gene. Mol Pharmacol 1998;54:687–94.

    PubMed  CAS  Google Scholar 

  172. Rahman Z, Gold SJ, Potenza MN, et al. Cloning and characterization of RGS9-2: A striatal-enriched alternatively spliced product of the RGS9 gene. J Neurosci 1999;19: 2016–26.

    PubMed  CAS  Google Scholar 

  173. Kovoor A, Seyffarth P, Ebert J, et al. D2 dopamine receptors colocalize regulator of G-protein signaling 9-2 (RGS9-2) via the RGS9 DEP domain, and RGS9 knock-out mice develop dyskinesias associated with dopamine pathways. J Neurosci 2005;25:2157–65.

    PubMed  CAS  Google Scholar 

  174. Rahman Z, Schwarz J, Gold SJ, et al. RGS9 modulates dopamine signaling in the basal ganglia. Neuron 2003;38:941.

    PubMed  CAS  Google Scholar 

  175. Geurts M, Hermans E, Maloteaux JM. Opposite modulation of regulators of G protein signalling-2 RGS2 and RGS4 expression by dopamine receptors in the rat striatum. Neurosci Lett 2002;333:146–50.

    PubMed  CAS  Google Scholar 

  176. Geurts M, Maloteaux JM, Hermans E. Altered expression of regulators of G-protein signaling (RGS) mRNAs in the striatum of rats undergoing dopamine depletion. Biochem Pharmacol 2003;66:1163–70.

    PubMed  CAS  Google Scholar 

  177. Gold SJ, Hoang CV, Potts BW, et al. RGS9 2 negatively modulates l-3,4-dihydroxyphenylalanine-Induced dyskinesia in experimental Parkinson’s disease. J Neurosci 2007;27:14338–48.

    PubMed  CAS  Google Scholar 

  178. Tekumalla PK, Calon F, Rahman Z, et al. Elevated levels of DeltaFosB and RGS9 in striatum in Parkinson’s disease. Biol Psychiatry 2001;50:813–6.

    PubMed  CAS  Google Scholar 

  179. Doucet JP, Nakabeppu Y, Bedard PJ, et al. Chronic alterations in dopaminergic neurotransmission produce a persistent elevation of deltaFosB-like protein(s) in both the rodent and primate striatum. Eur J Neurosci 1996;8:365–81.

    PubMed  CAS  Google Scholar 

  180. McClung CA, Ulery PG, Perrotti LI, Zachariou V, Berton O, Nestler EJ. DeltaFosB: a molecular switch for long-term adaptation in the brain. Brain Res Mol Brain Res 2004;132:146–54.

    PubMed  CAS  Google Scholar 

  181. Hornykiewicz O. Basic research on dopamine in Parkinson’s disease and the discovery of the nigrostriatal dopamine pathway: the view of an eyewitness. Neurodegener Dis 2008;5: 114–7.

    PubMed  CAS  Google Scholar 

  182. Marsden CD, Parkes JD. Success and problems of long-term levodopa therapy in Parkinson’s disease. Lancet 1977;309:345–9.

    Google Scholar 

  183. Asin KE, Bednarz L, Nikkel A, Perner R. Rotation and striatal c-fos expression after repeated, daily treatment with selective dopamine receptor agonists and levodopa. J Pharmacol Exp Ther 1995;273:1483–90.

    PubMed  CAS  Google Scholar 

  184. Rascol O, Nutt JG, Blin O, et al. Induction by dopamine D1 receptor agonist ABT-431 of dyskinesia similar to levodopa in patients with Parkinson disease. Arch Neurol 2001;58: 249–54.

    PubMed  CAS  Google Scholar 

  185. Blanchet PJ, Grondin R, Bédard PJ. Dyskinesia and wearing-off following dopamine D1 agonist treatment in drug-naive 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned primates. Mov Disord 1996;11:91–4.

    PubMed  CAS  Google Scholar 

  186. Blanchet PJ, Konitsiotis S, Chase TN. Motor response to a dopamine D3 receptor preferring agonist compared to apomorphine in levodopa-primed 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine monkeys. J Pharmacol Exp Ther 1997;283:794–9.

    PubMed  CAS  Google Scholar 

  187. Pearce RKB, Jackson M, Britton DR, Shiosaki K, Jenner P, Marsden CD. Actions of the D1 agonists A-77636 and A-86929 on locomotion and dyskinesia in MPTP-treated l-dopa-primed common marmosets. Psychopharmacology 1999;142:51–60.

    PubMed  CAS  Google Scholar 

  188. Rascol O, Blin O, Thalamas C, et al. ABT-431, a D1 receptor agonist prodrug, has efficacy in Parkinson’s disease. Ann Neurol 1999;45:736–41.

    PubMed  CAS  Google Scholar 

  189. Mercuri NB, Giorgio Bernardi G. The ‘magic’ of l-dopa: why is it the gold standard Parkinson’s disease therapy? Trends Pharmacol Sci 2005;26:341–4.

    PubMed  CAS  Google Scholar 

  190. FabbriniG, Brotchie JM, Grandas F, Nomoto M, Goet ZCG. Levodopa-induced dyskinesias. Mov Disord 2007;22:1379–89.

    PubMed  Google Scholar 

  191. Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor treatment of Parkinson’s disease: scientific rationale and clinical implications. Lancet Neurol 2006;5:677.

    PubMed  CAS  Google Scholar 

  192. Obeso JA, Rodriguez-Oroz MC, Rodrigue ZM, DeLong MR, Olanow CW. Pathophysiology of levodopa-induced dyskinesias in Parkinson’s disease: problems with the current model. Ann Neurol 2000;47:S22–32.

    Google Scholar 

  193. Crossman AR. A hypothesis on the pathophysiological mechanisms that underlie levodopa- or dopamine agonist-induced dyskinesia in Parkinson’s disease: Implications for future strategies in treatment. Mov Disord 1990;5:100–8.

    PubMed  CAS  Google Scholar 

  194. Kim DS, Palmiter RD, Cummins A, Gerfen CR. Reversal of supersensitive striatal dopamine D1 receptor signaling and extracellular signal-regulated kinase activity in dopamine-deficient mice. Neuroscience 2006;137:1381–8.

    PubMed  CAS  Google Scholar 

  195. Granata R, Wenning GK, Jolkkonen J, Jenner P, Marsden CD. Effect of repeated administration of dopamine agonists on striatal neuropeptide mRNA expression in rats with a unilateral nigral 6-hydroxydopamine lesion. J Neural Transm 1996;103:249–60.

    PubMed  CAS  Google Scholar 

  196. Tel BC, Zeng BY, Cannizzaro C, Pearce RK, Rose S, Jenner P. Alterations in striatal neuropeptide mRNA produced by repeated administration of l-DOPA, ropinirole or bromocriptine correlate with dyskinesia induction in MPTP-treated common marmosets. Neuroscience 2002;115:1047–58.

    PubMed  CAS  Google Scholar 

  197. Calon F, Birdi S, Rajput AH, Hornykiewicz O, Bédard PJ, Di Paolo T. Increase of preproenkephalin mRNA levels in the putamen of Parkinson disease patients with levodopa-induced dyskinesias. J Neuropathol Exp Neurol 2002;61:186–96.

    PubMed  CAS  Google Scholar 

  198. Gerfen CR, Engber TM, Mahan L, et al. D1 and D2 dopamine receptor-regulated gene expression of striatonigral and striatopallidal neurons. Science 1990;250:1429–32.

    PubMed  CAS  Google Scholar 

  199. Beaulieu J-M, Tirotta E, Sotnikova TD, et al. Regulation of Akt signaling by D2 and D3 dopamine receptors in vivo. J Neurosci 2007;27:881–5.

    PubMed  CAS  Google Scholar 

  200. Beaulieu JM, Gainetdinov RR, Caron MG. The Akt-GSK-3 signaling cascade in the actions of dopamine. Trends Pharmacol Sci 2007;28:166–72.

    PubMed  CAS  Google Scholar 

  201. NewDC, Wu K, Kwok AWS, Wong YH. G protein-coupled receptor-induced Akt activity in cellular proliferation and apoptosis. FEBS J 2007;274:6025–36.

    PubMed  CAS  Google Scholar 

  202. Rashid AJ, O’Dowd BF, Verma V, George SR. Neuronal Gq/11-coupled dopamine receptors: an uncharted role for dopamine. Trends Pharmacol Sci 2007;28:551–5.

    PubMed  CAS  Google Scholar 

  203. Brami-Cherrier K, Valjent E, Garcia M, Pages C, Hipskind RA, Caboche J. Dopamine induces a PI3-Kinase-Independent activation of Akt in striatal neurons: A new route to cAMP response element-binding protein phosphorylation. J Neurosci 2002;22:8911–21.

    PubMed  CAS  Google Scholar 

  204. Nielsen KM, Soghomonian JJ. Normalization of glutamate decarboxylase gene expression in the entopeduncular nucleus of rats with a unilateral 6-hydroxydopamine lesion correlates with increased gabaergic input following intermittent but not continuous levodopa. Neuroscience 2004;123:31.

    Google Scholar 

  205. Cenci MA, Lee CS, Björklund A.l-DOPA-induced dyskinesia in the rat is associated with striatal overexpression of prodynorphin- and glutamic acid decarboxylase mRNA. Eur J Neurosci 1998;10:2694–706.

    PubMed  CAS  Google Scholar 

  206. Hurley MJ, Jackson MJ, Smith LA, Rose S, Jenner P. Immunoautoradiographic analysis of NMDA receptor subunits and associated postsynaptic density proteins in the brain of dyskinetic MPTP-treated common marmosets. Eur J Neurosci 2005;21:3240–50.

    PubMed  CAS  Google Scholar 

  207. Calon F, Rajput AH, Hornykiewicz O, Bédard PJ, Di Paolo T. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson’s disease. Neurobiol Dis 2003;14:404–16.

    PubMed  CAS  Google Scholar 

  208. Dunah AW, Standaert DG. Dopamine D1 receptor-dependent trafficking of striatal NMDA glutamate receptors to the postsynaptic membrane. J Neurosci 2001;21:5546–58.

    PubMed  CAS  Google Scholar 

  209. Dunah AW, Sirianni AC, Fienberg AA, Bastia E, Schwarzschild MA, Standaert DG. Dopamine D1-dependent trafficking of striatal N-methyl-D-aspartate glutamate receptors requires Fyn protein tyrosine kinase but not DARPP-32. Mol Pharmacol 2004;65:121–9.

    PubMed  CAS  Google Scholar 

  210. Hallett PJ, Spoelgen R, Hyman BT, Standaert DG, Dunah AW. Dopamine D1 activation potentiates striatal NMDA receptors by tyrosine phosphorylation-dependent subunit trafficking. J Neurosci 2006;26:4690–700.

    PubMed  CAS  Google Scholar 

  211. Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Movement Disorders 2001;16:448–58.

    PubMed  CAS  Google Scholar 

  212. Sgambato-Faure V, Buggia V, Gilbert F, Levesque D, Benabid AL, Berger F. Coordinated and spatial upregulation of arc in striatonigral neurons correlates with l-dopa-induced behavioral sensitization in dyskinetic rats. J Neuropathol Exp Neurol 2005;64:936–47.

    PubMed  CAS  Google Scholar 

  213. Cenci MA, Whishaw IQ, Schallert T. Animal models of neurological deficits: how relevant is the rat? Nat Rev Neurosci 2002;3:574–9.

    PubMed  CAS  Google Scholar 

  214. Carta M, Lindgren HS, Lundblad M, Stancampiano R, Fadda F, Cenci MA. Role of striatal l-DOPA in the production of dyskinesia in 6-hydroxydopamine lesioned rats. J Neurochem 2006;96:1718–28.

    PubMed  CAS  Google Scholar 

  215. Guigoni C, Dovero S, Aubert I, et al. Levodopa-induced dyskinesia in MPTP-treated macaques is not dependent on the extent and pattern of nigrostrial lesioning. Eur J Neurosci 2005;22:283–7.

    PubMed  Google Scholar 

  216. Putterman DB, Munhall AC, Kozell LB, Belknap JK, Johnson SW. Evaluation of levodopa dose and magnitude of dopamine depletion as risk factors for levodopa-induced dyskinesia in a rat model of Parkinson’s disease. J Pharmacol Exp Ther 2007;323:277–84.

    PubMed  CAS  Google Scholar 

  217. Hauser RA, McDermott MP, Messing S. Factors associated with the development of motor fluctuations and dyskinesias in Parkinson disease. Arch Neurol 2006;63:1756–60.

    PubMed  Google Scholar 

  218. Schrag A, Quinn N. Dyskinesias and motor fluctuations in Parkinson’s disease: a community-based study. Brain 2000;123:2297–305.

    PubMed  Google Scholar 

  219. Winkle RC, Kirik D, Björklund A, Cenci MA.L-DOPA-induced dyskinesia in the intrastriatal 6-hydroxydopamine model of Parkinson’s disease: relation to motor and cellular parameters of nigrostriatal function. Neurobiol Dis 2002;10:165–86.

    Google Scholar 

  220. Pavon N, Martin AB, Mendialdua A, Moratalla R. ERK phosphorylation and FosB expression are associated with l-DOPA-induced dyskinesia in hemiparkinsonian mice. Biol Psychiatry 2006;59:64–74.

    PubMed  CAS  Google Scholar 

  221. Hardingham GH, Fukunaga Y, Hilmar Bading H. Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat Neurosci 2002;5:405–14.

    PubMed  CAS  Google Scholar 

  222. Andersson M, Hilbertson A, Cenci MA. Striatal fosB expression is causally linked with l-DOPA-induced abnormal involuntary movements and the associated upregulation of striatal prodynorphin mRNA in a rat model of Parkinson’s disease. Neurobiol Dis 1999;6:461–74.

    PubMed  CAS  Google Scholar 

  223. Cenci MA. Transcription factors involved in the pathogenesis of l-DOPA-induced dyskinesia in a rat model of Parkinson’s disease. Amino Acids 2002;23:105–9.

    PubMed  CAS  Google Scholar 

  224. Zachariou V, Sgambato-Faure V, Sasaki T, et al. Phosphorylation of DARPP-32 at Threonine-34 is required for cocaine action. Neuropsychopharmacology 2006;31:555–62.

    PubMed  CAS  Google Scholar 

  225. Hiroi N, Fienberg AA, Haile CN, et al. Neuronal and behavioural abnormalities in striatal function in DARPP-32-mutant mice. Eur J Neurosci 1999;11:1114–8.

    PubMed  CAS  Google Scholar 

  226. Hiroi N, Brown JR, Haile CN, Ye H, Greenberg ME, Nestler EJ. FosB mutant mice: loss of chronic cocaine induction of Fos-related proteins and heightened sensitivity to cocaine’s psychomotor and rewarding effects. Proc Natl Acad Sci USA 1997;94:10397–402.

    PubMed  CAS  Google Scholar 

  227. Kelz MB, Chen J, Carlezon WA, et al. Expression of the transcription factor DeltaFosB in the brain controls sensitivity to cocaine. Nature 1999;401:272.

    PubMed  CAS  Google Scholar 

  228. Tzingounis AV, Nicoll RA. Arc/Arg3.1: linking gene expression to synaptic plasticity and memory. Neuron 2006;52:403–7.

    PubMed  CAS  Google Scholar 

  229. Chowdhury S, Shepherd JD, Okuno H, et al. Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA receptor trafficking. Neuron 2006;52:445.

    PubMed  CAS  Google Scholar 

  230. Shepherd JD, Rumbaugh G, Wu J, et al. Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron 2006;52:475.

    PubMed  CAS  Google Scholar 

  231. Rial Verde EM, Lee-Osbourne J, Worley PF, Malinow R, Cline HT. Increased expression of the immediate-early gene Arc/Arg3.1 reduces AMPA receptor-mediated synaptic transmission. Neuron 2006;52:461.

    PubMed  Google Scholar 

  232. Bibbiani F, Oh JD, Kielaite A, Collins MA, Smith C, Chase TN. Combined blockade of AMPA and NMDA glutamate receptors reduces levodopa-induced motor complications in animal models of PD. Exp Neurol 2005;196:422.

    PubMed  CAS  Google Scholar 

  233. Shiraishi-Yamaguchi Y, Furuichi T. The Homer family proteins. Genome Biol 2007;8:206.

    PubMed  Google Scholar 

  234. Kammermeier PJ, Worley PF. Homer 1a uncouples metabotropic glutamate receptor 5 from postsynaptic effectors. Proc Natl Acad Sci USA 2007;104:6055–60.

    PubMed  CAS  Google Scholar 

  235. Tappe A, Kuner R. Regulation of motor performance and striatal function by synaptic scaffolding proteins of the Homer1 family. Proc Natl Acad Sci USA 2006;103:774–9.

    PubMed  CAS  Google Scholar 

  236. Gerfen CR. The neostriatal mosaic: multiple levels of compartmental organization. Trends Neurosci 1992;15:133–9.

    PubMed  CAS  Google Scholar 

  237. Prensa L, Parent A. The nigrostriatal pathway in the rat: A single-axon study of the relationship between dorsal and ventral tier nigral neurons and the striosome/matrix striatal compartments. J Neurosci 2001;21:7247–60.

    PubMed  CAS  Google Scholar 

  238. Joel D, Weiner I. The connections of the dopaminergic system with the striatum in rats and primates: an analysis with respect to the functional and compartmental organization of the striatum. Neuroscience 2000;96:451.

    PubMed  CAS  Google Scholar 

  239. Saka E, Elibol B, Erdem S, Dalkara T. Compartmental changes in expression of c-Fos and FosB proteins in intact and dopamine-depleted striatum after chronic apomorphine treatment. Brain Res 1999;825:104–14.

    PubMed  CAS  Google Scholar 

  240. Bonuccelli U.Comparing dopamine agonists in Parkinson’s disease. Curr Opin Neurol 2003;16(Suppl 1):S13–9.

    PubMed  Google Scholar 

  241. Bonuccelli U, Pavese N. Dopamine agonists in the treatment of Parkinson’s disease. Expert Rev Neurother 2006;6:81–9.

    PubMed  CAS  Google Scholar 

  242. Foley P, Gerlach M, Double KL, Riederer P. Dopamine receptor agonists in the therapy of Parkinson’s disease. J Neural Transm 2004;111:1375–446.

    PubMed  CAS  Google Scholar 

  243. BibbianiF, Costantini LC, Pate lR, Chase TN. Continuous dopaminergic stimulation reduces risk of motor complications in parkinsonian primates. Exp Neurol 2005;192:7378.

    Google Scholar 

  244. Blanchet PJ, Calon F, Martel JC, et al. Continuous administration decreases and pulsatile administration increases behavioral sensitivity to a novel dopamine D2 agonist (U-91356a) in MPTP- exposed monkeys. J Pharmacol Exp Ther 1995;272:854–9.

    PubMed  CAS  Google Scholar 

  245. Maratos EC, Jackson MJ, Pearce RK, Cannizzaro C, Jenner P. Both short- and long-acting D-1/D-2 dopamine agonists induce less dyskinesia than l-DOPA in the MPTP-lesioned common marmoset (Callithrix jacchus). Exp Neurol 2003;179:90–102.

    PubMed  CAS  Google Scholar 

  246. Delfino M, Kalisch R, Czisch M, et al. Mapping the effects of three dopamine agonists with different dyskinetogenic potential and receptor selectivity using pharmacological functional magnetic resonance imaging. Neuropsychopharmacology 2007;32:1911.

    PubMed  CAS  Google Scholar 

  247. Delfino MA, Stefano AV, Ferrario JE, Taravini IRE, Murer MG, Gershanik OS. Behavioral sensitization to different dopamine agonists in a parkinsonian rodent model of drug-induced dyskinesias. Behav Brain Res 2004;152:297.

    PubMed  CAS  Google Scholar 

  248. Blanchet P, Bedard PJ, Britton DR, Kebabian JW. Differential effect of selective D-1 and D-2 dopamine receptor agonists on levodopa-induced dyskinesia in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- exposed monkeys. J Pharmacol Exp Ther 1993;267:275–9.

    PubMed  CAS  Google Scholar 

  249. Bianchi BR, Miller TR, Stashko MA, et al. Persistent activation of the dopamine D1 receptor contributes to prolonged receptor desensitization: Studies with A-77636. J Pharmacol Exp Ther 1996;276:1022–9.

    PubMed  Google Scholar 

  250. Blanchet PJ, Grondin R, Bedard PJ, Shiosaki K, Britton DR. Dopamine D1 receptor desensitization profile in MPTP-lesioned primates. Eur J Pharmacol 1996;309:13.

    PubMed  CAS  Google Scholar 

  251. Nutt JG. Continuous dopaminergic stimulation: Is it the answer to the motor complications of Levodopa? Mov Disord 2007;22:1–9.

    PubMed  Google Scholar 

  252. Stocchi F, Olanow CW. Continuous dopaminergic stimulation in early and advanced Parkinson’s disease. Neurology 2004;62(Suppl 1):S56–63.

    PubMed  Google Scholar 

  253. Olanow CW, Obeso JA, Stocchi F. Drug insight: Continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Nat Clin Pract Neurol 2006;2:382–92.

    PubMed  CAS  Google Scholar 

  254. Stocchi F, Vacca L, Ruggieri S, Olanow CW. Intermittent vs continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol 2005;62:905–10.

    PubMed  Google Scholar 

  255. Ravenscroft P, Chalon S, Brotchie JM, Crossman AR. Ropinirole versus l-DOPA effects on striatal opioid peptide precursors in a rodent model of Parkinson’s disease: implications for dyskinesia. Exp Neurol 2004;185:36–46.

    PubMed  CAS  Google Scholar 

  256. Jenner P. A novel dopamine agonist for the transdermal treatment of Parkinson’s disease. Neurology 2005;65:S3–S5.

    Google Scholar 

  257. LeWitt PA, Lyons KE, Pahwa R, on behalf of the SPSG. Advanced Parkinson disease treated with rotigotine transdermal system: PREFER Study. Neurology 2007;68:1262–7.

    PubMed  CAS  Google Scholar 

  258. Winkler JD, Weiss B. Reversal of supersensitive apomorphine-induced rotational behavior in mice by continuous exposure to apomorphine. J Pharmacol Exp Ther 1986;238:242–7.

    PubMed  CAS  Google Scholar 

  259. Linazasoro G, Obeso JA, Gómez JC, Martínez M, Antonini A, Leenders KL. Modification of dopamine D2 receptor activity by pergolide in Parkinson’s disease: An in vivo study by PET. Clin Neuropharmacol 1999;22:277–80.

    PubMed  CAS  Google Scholar 

  260. Goulet M, Grondin R, Morissette M, et al. Regulation by chronic treatment with cabergoline of dopamine D1 and D2 receptor levels and their expression in the striatum of Parkinsonian-monkeys. Progr Neuro-Psychopharmacol Biol Psychiatry 2000;24:607.

    CAS  Google Scholar 

  261. Muriel MP, Bernard V, Levey AI, et al. Levodopa induces a cytoplasmic localization of D1 dopamine receptors in striatal neurons in Parkinson’s disease. Ann Neurol 1999;46:103–11.

    PubMed  CAS  Google Scholar 

  262. Guigoni C, Doudnikoff E, Li Q, Bloch B, Bezard E. Altered D1 dopamine receptor trafficking in parkinsonian and dyskinetic non-human primates. Neurobiol Dis 2007;26:452.

    PubMed  CAS  Google Scholar 

  263. Kim OJ, Gardner BR, Williams DB, et al. The role of phosphorylation in D1 dopamine receptor desensitization: evidence for a novel mechanism of arrestin association. J Biol Chem 2004;279:7999–8010.

    PubMed  CAS  Google Scholar 

  264. Kim KM, Valenzano KJ, Robinson SR, Yao WD, Barak LS, Caron MG. Differential regulation of the dopamine D2 and D3 receptors by G protein-coupled receptor kinases and beta-arrestins. J Biol Chem 2001;276:37409–14.

    PubMed  CAS  Google Scholar 

  265. Kim KM, Gainetdinov RR, Laporte SA, Caron MG, Barak LS. G protein-coupled receptor kinase regulates dopamine D3 receptor signaling by modulating the stability of a receptor-filamin-beta-arrestin complex. A case of autoreceptor regulation. J Biol Chem 2005;280:12774–80.

    PubMed  CAS  Google Scholar 

  266. Cho DI, Beom S, Van Tol HH, Caron MG, Kim KM. Characterization of the desensitization properties of five dopamine receptor subtypes and alternatively spliced variants of dopamine D2 and D4 receptors. Biochem Biophys Res Commun 2006;350:634–40.

    PubMed  CAS  Google Scholar 

  267. Szczypka MS, Rainey MA, Kim DS, et al. Feeding behavior in dopamine-deficient mice. Proc Natl Acad Sci USA 1999;96:12138–43.

    PubMed  CAS  Google Scholar 

  268. Steige RM. Constant dopaminergic stimulation by transdermal delivery of dopaminergic drugs: a new treatment paradigm in Parkinson’s disease. Eur J Neurol 2008;15:6–15.

    Google Scholar 

  269. Grace AA. Phasic versus tonic dopamine release and the modulation of dopamine system responsivity: A hypothesis for the etiology of schizophrenia. Neuroscience 1991;41:1.

    PubMed  CAS  Google Scholar 

  270. West AR, Grace AA. Opposite influences of endogenous dopamine D1 and D2 receptor activation on activity states and electrophysiological properties of striatal neurons: studies combining in vivo intracellular recordings and reverse microdialysis. J Neurosci 2002;22:294–304.

    PubMed  CAS  Google Scholar 

  271. Grace AA. The tonic/phasic model of dopamine system regulation and its implications for understanding alcohol and psychostimulant craving. Addiction 2000;95(Suppl 2):S119–S28.

    Google Scholar 

  272. Schultz W. Predictive reward signal of dopamine neurons. J Neurophysiol 1998;80:1–27.

    PubMed  CAS  Google Scholar 

  273. de la Fuente-Fernández R, Sossi V, Huang Z, et al. Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: implications for dyskinesias. Brain 2004;127:2747–54.

    PubMed  Google Scholar 

  274. Miller DW, Abercrombie ED. Role of high-affinity dopamine uptake and impulse activity in the appearance of extracellular dopamine in striatum after administration of exogenous l-DOPA: studies in intact and 6-hydroxydopamine-treated rats. J Neurochem 1999;72: 1516–22.

    PubMed  CAS  Google Scholar 

  275. Tedroff J, Pedersen M, Aquilonius SM, Hartvig P, Jacobsson G, Långström B. Levodopa-induced changes in synaptic dopamine in patients with Parkinson’s disease as measured by [11C]raclopride displacement and PET. Neurology 1996;46:1430–6.

    PubMed  CAS  Google Scholar 

  276. Brooks DJ, Sagar H. Entacapone is beneficial in both fluctuating and non-fluctuating patients with Parkinson’s disease: a randomised, placebo controlled, double blind, six month study. J Neurol Neurosurg Psychiatry 2003;74:1071–9.

    PubMed  CAS  Google Scholar 

  277. Ahmed MR, Bychkov E, Gurevich EV. Lentivirus-mediated overexpression of GRK3 and 5 suppresses behavioral sensitization to l-DOPA in the 6-OHDA rat model of Parkinson’s disease. Soc Neurosci Abstr San Diego, 2007.

    Google Scholar 

  278. Maratos EC, Jackson MJ, Pearce RKB, Jenner P. Antiparkinsonian activity and dyskinesia risk of ropinirole and l-DOPA combination therapy in drug naive MPTP-lesioned common marmosets (Callithrix jacchus). Mov Disord 2001;16:631–41.

    PubMed  CAS  Google Scholar 

  279. Verhagen Metman L, Locatelli ER, Bravi D, Mouradian MM, Chase TN. Apomorphine responses in Parkinson’s disease and the pathogenesis of motor complications. Neurology 1997;48:369–72.

    PubMed  CAS  Google Scholar 

  280. Grandas F, Gancher ST, Rodriguez M, Lera G, Nutt JG, Obeso JA. Differences in the motor response to apomorphine between untreated and fluctuating patients with Parkinson’s disease. Clin Neuropharmacol 1992;15:13–8.

    PubMed  CAS  Google Scholar 

  281. Bravi D, Mouradian MM, Roberts JW, Davis TL, Sohn YH, Chase TN. Wearing-off fluctuations in Parkinson’s disease: Contribution of postsynaptic mechanisms. Ann Neurol 1994;36:27–31.

    PubMed  CAS  Google Scholar 

  282. Colosimo C, Merello M., Hughes AJ, Sieradzan K, Lees AJ. Motor response to acute dopaminergic challenge with apomorphine and levodopa in Parkinson’s disease: implications for the pathogenesis of the on-off phenomenon. J Neurol Neurosurg Psychiatry 1996;60:634–7.

    PubMed  CAS  Google Scholar 

  283. Harder S, Baas H. Concentration-response relationship of levodopa in patients at different stages of Parkinson’s disease. Clin Pharmacol Ther 1998;64:183–91.

    PubMed  CAS  Google Scholar 

  284. Nutt JG, Carter JH, Lea ES, Sexton GJP. Evolution of the response to levodopa during the first 4 years of therapy. Ann Neurol 2002;51:686–93.

    PubMed  CAS  Google Scholar 

  285. Voon V, Potenza MN, Thomsen T. Medication-related impulse control and repetitive behaviors in Parkinson’s disease. Curr Opin Neurol 2007;20:484–92.

    PubMed  Google Scholar 

  286. Nirenberg MJ, Waters C. Compulsive eating and weight gain related to dopamine agonist use. Mov Disord 2006;21:524–9.

    PubMed  Google Scholar 

  287. Weintraub D, Potenza MN. Impulse control disorders in Parkinson’s disease. Curr Neurol Neurosci Rep 2006;6:302–6.

    PubMed  Google Scholar 

  288. Freed CR, Greene PE, Breeze RE, et al. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Engl J Med 2001;344:710–9.

    PubMed  CAS  Google Scholar 

  289. Hagell P, Piccini P, Björklund A, et al. Dyskinesias following neural transplantation in Parkinson’s disease. Nat Neurosci 2002;5:627–8.

    PubMed  CAS  Google Scholar 

  290. Olanow WC, Goetz CG, Kordower JH, et al. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson’s disease. Ann Neurol 2003;54:403–14.

    PubMed  Google Scholar 

  291. OhJD, Geller AI, Zhang G, Chase TN. Gene transfer of constitutively active protein kinase C into striatal neurons accelerates onset of levodopa-induced motor response alterations in parkinsonian rats. Brain Res 2003;971:18–30.

    PubMed  CAS  Google Scholar 

  292. Mazzella L, Yahr MD, Marinelli L, Huang N, Moshier E, Di Rocco A. Dyskinesias predict the onset of motor response fluctuations in patients with Parkinson’s disease on l-dopa monotherapy. Parkinsonism Relat Disord 2005;11:151–5.

    PubMed  CAS  Google Scholar 

  293. Bové J, Serrats J, Mengod G, Cortés R, Aguilar E, Marin C. Reversion of levodopa-induced motor fluctuations by the A2A antagonist CSC is associated with an increase in striatal preprodynorphin mRNA expression in 6-OHDA-lesioned rats. Synapse 2006;59:435–44.

    PubMed  Google Scholar 

  294. Wessell RH, Ahmed SM, Menniti FS, Dunbar GL, Chase TN, Oh JD. NR2B selective NMDA receptor antagonist CP-101,606 prevents levodopa-induced motor response alterations in hemi-parkinsonian rats. Neuropharmacology 2004;47:184–94.

    PubMed  CAS  Google Scholar 

  295. Bibbiani F, Oh JD, Petzer JP, et al. A2A antagonist prevents dopamine agonist-induced motor complications in animal models of Parkinson’s disease. Exp Neurol 2003;184: 285–94.

    PubMed  CAS  Google Scholar 

  296. Oh JD, Bibbiani F, Chase TN. Quetiapine attenuates levodopa-induced motor complications in rodent and primate parkinsonian models. Exp Neurol 2002;177:557–64.

    PubMed  CAS  Google Scholar 

  297. Hurley MJ, Jenner P. What has been learnt from study of dopamine receptors in Parkinson’s disease? Pharmacol Ther 2006;111:715.

    PubMed  CAS  Google Scholar 

  298. Meador-Woodruff JH, Damask SP, Watson SJJ. Differential expression of autoreceptors in the ascending dopamine systems of the human brain. Proc Natl Acad Sci USA 1994;91:8297–301.

    PubMed  CAS  Google Scholar 

  299. Mrzljak L, Bergson C, Pappy M, Huff R, Levenson R, Goldman-Rakic PS. Localization of dopamine D4 receptors in GABAergic neurons of the primate brain. Nature 1996;381:245–8.

    PubMed  CAS  Google Scholar 

  300. Svingos AL, Periasamy S, Pickel VM. Presynaptic dopamine D(4) receptor localization in the rat nucleus accumbens shell. Synapse 2000;36:222–32.

    PubMed  CAS  Google Scholar 

  301. Caillé I, Dumartin B, Bloch B. Ultrastructural localization of D1 dopamine receptor immunoreactivity in rat striatonigral neurons and its relation with dopaminergic innervation. Brain Res 1996;730:17–31.

    PubMed  Google Scholar 

  302. Beischlag TV, Marchese A, Meador-Woodruff JH, et al. The human dopamine D5 receptor gene: cloning and characterization of the 5'-flanking and promoter region. Biochemistry 1995;34:5960–70.

    PubMed  CAS  Google Scholar 

  303. Moore DJ, West AB, Dawson VL, Dawson TM. Molecular pathophysiology of Parkinson’s disease. Annu Rev Neurosci 2005;28:57–87.

    PubMed  CAS  Google Scholar 

  304. Mytilineou C, Han SK, Cohen G. Toxic and protective effects of l-dopa on mesencephalic cell cultures. J Neurochem 1993;61:1470–8.

    PubMed  CAS  Google Scholar 

  305. Michel PP, Hefti F. Toxicity of 6-hydroxydopamine and dopamine for dopaminergic neurons in culture. J Neurosci Res 1990;26:428–35.

    PubMed  CAS  Google Scholar 

  306. Steece-Collier K, Collier TJ, Sladek CD, Sladek JRJ. Chronic levodopa impairs morphological development of grafted embryonic dopamine neurons. Exp Neurol 1990;110: 201–8.

    PubMed  CAS  Google Scholar 

  307. Mena MA, Pardo B, Paino CL, De Yebenes JG. Levodopa toxicity in foetal rat midbrain neurones in culture: modulation by ascorbic acid. Neuroreport 1993;4:438–40.

    PubMed  CAS  Google Scholar 

  308. Pardo B, Mena MA, Casarejos MJ, Paíno CL, De Yébenes JG. Toxic effects of l-DOPA on mesencephalic cell cultures: protection with antioxidants. Brain Res 1995;682:133–43.

    PubMed  CAS  Google Scholar 

  309. Han SK, Mytilineou C, Cohen G. l-DOPA up-regulates glutathione and protects mesencephalic cultures against oxidative stress. J Neurochem 1996;66:501–10.

    PubMed  CAS  Google Scholar 

  310. Ling ZD, Pieri SC, Carvey PM. Comparison of the neurotoxicity of dihydroxyphenylalanine stereoisomers in cultured dopamine neurons. Clin Neuropharmacol 1996;19:360–5.

    PubMed  CAS  Google Scholar 

  311. MenaMA, Casarejos MJ, Carazo A, Paíno CL, García de Yébenes J. Glia protect fetal midbrain dopamine neurons in culture from l-DOPA toxicity through multiple mechanisms. J Neural Transm 1997;104:317–28.

    PubMed  CAS  Google Scholar 

  312. Mena MA, Davila V, Sulzer D. Neurotrophic effects of l-DOPA in postnatal midbrain dopamine neuron/cortical astrocyte cocultures. J Neurochem 1997;69:1398–408.

    PubMed  CAS  Google Scholar 

  313. Mena MA, Davila V, Bogaluvsky J, Sulzer D. A synergistic neurotrophic response to l-dihydroxyphenylalanine and nerve growth factor. Mol Pharmacol 1998;54:678–86.

    PubMed  CAS  Google Scholar 

  314. Chen J, Wersinger C, Sidhu A. Chronic stimulation of D1 dopamine receptors in human SK-N-MC neuroblastoma cells induces nitric-oxide synthase activation and cytotoxicity. J Biol Chem 2003;278:28089–100.

    PubMed  CAS  Google Scholar 

  315. Chen J, Rusnak M, Luedtke RR, Sidhu A. D1 Dopamine Receptor Mediates Dopamine-induced Cytotoxicity via the ERK Signal Cascade. J Biol Chem 2004;279:39317–30.

    PubMed  CAS  Google Scholar 

  316. Mytilineou C, Walker RH, JnoBaptiste R, Olanow CW. Levodopa is toxic to dopamine neurons in an in vitro but not an in vivo model of oxidative stress. J Pharmacol Exp Ther 2003;304:792–800.

    PubMed  CAS  Google Scholar 

  317. Murer M, Dziewczapolski G, Menalled LB, et al. Chronic levodopa is not toxic for remaining dopamine neurons, but instead promotes their recovery, in rats with moderate nigrostriatal lesions. Ann Neurol 1998;43:561–75.

    PubMed  CAS  Google Scholar 

  318. Datla KP, Blunt SB, Dexter DT. Chronic l-DOPA administration is not toxic to the remaining dopaminergic nigrostriatal neurons, but instead may promote their functional recovery, in rats with partial 6-OHDA or FeCl3 nigrostriatal lesions. Mov Disord 2001;16:424–34.

    PubMed  CAS  Google Scholar 

  319. Fahn S. A new look at levodopa based on the ELLDOPA study. J Neural Transm Suppl 2006;70:419–26.

    PubMed  CAS  Google Scholar 

  320. Fahn S, the Parkinson Study Group. Does levodopa slow or hasten the rate of progression of Parkinson’s disease? J Neurol 2005;252(Suppl 4):IV37–IV42.

    Google Scholar 

  321. Group PS. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002;287:1653–61.

    Google Scholar 

  322. Whone AL, Watts RL, Stoessl AJ, et al. Slower progression of Parkinson’s disease with ropinirole versus levodopa: The REAL-PET study. Ann Neurol 2003;54:93–101.

    PubMed  CAS  Google Scholar 

  323. Scheller D, Chan P, Li Q, et al. Rotigotine treatment partially protects from MPTP toxicity in a progressive macaque model of Parkinson’s disease. Exp Neurol 2007;203:415–22.

    PubMed  CAS  Google Scholar 

  324. Double KL, Halliday GM, Henderson J, et al. The dopamine receptor agonist lisuride attenuates iron-mediated dopaminergic neurodegeneration. Exp Neurol 2003;184:530–5.

    PubMed  CAS  Google Scholar 

  325. JoyceJN, Steve Presgraves S, Renisha L, et al. Neuroprotective effects of the novel D3/D2 receptor agonist and antiparkinson agent, S32504, in vitro against 1-methyl-4-phenylpyridinium (MPP+) and in vivo against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): a comparison to ropinirole. Exp Neurol 2003;184:393–407.

    PubMed  CAS  Google Scholar 

  326. JoyceJN, Woolsey C, Ryoo H, Borwege S, Diane Hagner D. Low dose pramipexole is neuroprotective in the MPTP mouse model of Parkinson’s disease, and downregulates the dopamine transporter via the D3 receptor. BMC Biol 2004;2:22.

    PubMed  Google Scholar 

  327. Iida M, Miyazaki I, Tanaka K, Kabuto H, Iwata-Ichikawa E, Ogawa N. Dopamine D2 receptor-mediated antioxidant and neuroprotective effects of ropinirole, a dopamine agonist. Brain Res 1999;838:51–9.

    PubMed  CAS  Google Scholar 

  328. Anderson DW, Neavin T, Smith JA, Schneider JS. Neuroprotective effects of pramipexole in young and aged MPTP-treated mice. Brain Res 2001;905:44–53.

    PubMed  CAS  Google Scholar 

  329. Iravani MM, Haddon CO, Cooper JM, Jenner P, Schapira AH. Pramipexole protects against MPTP toxicity in non-human primates. J Neurochem 2006;96:1315–21.

    PubMed  CAS  Google Scholar 

  330. Vu TQ, Ling ZD, Ma SY, et al. Pramipexole attenuates the dopaminergic cell loss induced by intraventricular 6-hydroxydopamine. J Neural Transm 2000;107:159–76.

    PubMed  CAS  Google Scholar 

  331. Takashimaa H, Tsujihataa M, Kishikawab M, Freed WJ. Bromocriptine protects dopaminergic neurons from levodopa-induced toxicity by stimulating D2 receptors. Exp Neurol 1999;159:98–104.

    Google Scholar 

  332. Sawada H, Ibi M, Kihara T, et al. Dopamine D2-type agonists protect mesencephalic neurons from glutamate neurotoxicity: Mechanisms of neuroprotective treatment against oxidative stress. Ann Neurol 1998;44:110–9.

    PubMed  CAS  Google Scholar 

  333. Presgraves SP, Borwegea S, Millan MJ, Joyce JN. Involvement of dopamine D2/D3 receptors and BDNF in the neuroprotective effects of S32504 and pramipexole against 1-methyl-4-phenylpyridinium in terminally differentiated SH-SY5Y cells. Exp Neurol 2004;190:157–70.

    PubMed  CAS  Google Scholar 

  334. Yu Y, Wang JR, Sun PH, et al. Neuroprotective effects of atypical D1 receptor agonist SKF83959 are mediated via D1 receptor-dependent inhibition of glycogen synthase kinase-3 beta and a receptor-independent anti-oxidative action. J Neurochem 2008;104:946–56.

    PubMed  CAS  Google Scholar 

  335. Joyce JN, Millan MJ. Dopamine D3 receptor agonists for protection and repair in Parkinson’s disease. Curr Opin Pharmacol 2007;7:100–5.

    PubMed  CAS  Google Scholar 

  336. Ramirez AD, Wong SK-F, Menniti FS. Pramipexole inhibits MPTP toxicity in mice by dopamine D3 receptor dependent and independent mechanisms. Eur J Pharmacol 2003;475:29–35.

    PubMed  CAS  Google Scholar 

  337. Du F, Li R, Huang Y, Li X, Le W. Dopamine D3 receptor-preferring agonists induce neurotrophic effects on mesencephalic dopamine neurons. Eur J Neurosci 2005;22:2422–30.

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Eugenia V. Gurevich .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2010 Humana Press, a part of Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Gurevich, E.V., Gurevich, V.V. (2010). Dopamine Receptors and the Treatment of Parkinson’s Disease. In: Neve, K. (eds) The Dopamine Receptors. The Receptors. Humana Press, Totowa, NJ. https://doi.org/10.1007/978-1-60327-333-6_18

Download citation

Publish with us

Policies and ethics