Skip to main content
Top
Published in: Journal of Neurology 12/2014

01-12-2014 | Review

Dopamine receptor mapping with PET imaging in Parkinson’s disease

Authors: Flavia Niccolini, Paul Su, Marios Politis

Published in: Journal of Neurology | Issue 12/2014

Login to get access

Abstract

Parkinson’s disease (PD) is a chronic neurodegenerative disorder characterised pathologically by the loss of dopaminergic neurons in the substantia nigra pars compacta. These neurons project to the striatum, and their loss leads to alterations in the activity of the neural circuits that regulate movement. The striatal output of the circuit related to the control of movement is mediated by two pathways: the direct striatal pathway, which is mediated through facilitation of D1 receptors, and the indirect striatal pathway, mediated through D2 receptors. Positron emission tomography (PET) molecular imaging is a powerful in vivo technique in which using selective dopaminergic radioligands has been employed to investigate the dopaminergic system in humans. In this article we aim to review the role of PET imaging in understanding the postsynaptic dopaminergic mechanisms in PD. PET studies have allowed us to gain important insights into the functions of the dopaminergic system, the mechanisms of drug-induced motor and non-motor complications, and the placebo effect in PD.
Literature
3.
go back to reference Jellinger KA (1991) Pathology of Parkinson’s disease. Mol Chem Neuropathol 3:153–197CrossRef Jellinger KA (1991) Pathology of Parkinson’s disease. Mol Chem Neuropathol 3:153–197CrossRef
5.
go back to reference Levy G, Tang MX, Cote LJ, Louise ED, Alfaro B, Mejia H, Stern Y, Marder K (2000) Motor impairment in Parkinson’s disease: relationship to incident dementia and age. Neurology 55:539–544CrossRefPubMed Levy G, Tang MX, Cote LJ, Louise ED, Alfaro B, Mejia H, Stern Y, Marder K (2000) Motor impairment in Parkinson’s disease: relationship to incident dementia and age. Neurology 55:539–544CrossRefPubMed
6.
go back to reference Chesselet MF, Delfs JM (1999) Basal ganglia and movement disorders: an update. Trends Neurosci 19:417–422CrossRef Chesselet MF, Delfs JM (1999) Basal ganglia and movement disorders: an update. Trends Neurosci 19:417–422CrossRef
7.
go back to reference Hamani C, Lozano AM (2003) Physiology and pathophysiology of Parkinson’s disease. Ann NY Acad Sci 991:15–21CrossRefPubMed Hamani C, Lozano AM (2003) Physiology and pathophysiology of Parkinson’s disease. Ann NY Acad Sci 991:15–21CrossRefPubMed
8.
go back to reference Politis M, Piccini P (2012) Positron emission tomography imaging in neurological disorders. J Neurol 259(9):1769–1780CrossRefPubMed Politis M, Piccini P (2012) Positron emission tomography imaging in neurological disorders. J Neurol 259(9):1769–1780CrossRefPubMed
10.
go back to reference Halldin C, Stone-Elander S, Farde L, Ehrin E, Fasth KJ, Långström B, Sedvall G (1986) Preparation of 11C-labelled SCH 23390 for the in vivo study of dopamine D-1 receptors using positron emission tomography. Int J Rad Appl Instrum A 37(10):1039–1043CrossRefPubMed Halldin C, Stone-Elander S, Farde L, Ehrin E, Fasth KJ, Långström B, Sedvall G (1986) Preparation of 11C-labelled SCH 23390 for the in vivo study of dopamine D-1 receptors using positron emission tomography. Int J Rad Appl Instrum A 37(10):1039–1043CrossRefPubMed
11.
go back to reference Slifstein M, Kegeles LS, Gonzales R, Frankle WG, Xu X, Laruelle M, Abi-Dargham A (2007) [11C]NNC 112 selectivity for dopamine D1 and serotonin 5-HT(2A) receptors: a PET study in healthy human subjects. J Cereb Blood Flow Metab 27(10):1733–1741CrossRefPubMed Slifstein M, Kegeles LS, Gonzales R, Frankle WG, Xu X, Laruelle M, Abi-Dargham A (2007) [11C]NNC 112 selectivity for dopamine D1 and serotonin 5-HT(2A) receptors: a PET study in healthy human subjects. J Cereb Blood Flow Metab 27(10):1733–1741CrossRefPubMed
12.
go back to reference Chou YH, Karlsson P, Halldin C, Olsson H, Farde L (1999) A PET study of D(1)-like dopamine receptor ligand binding during altered endogenous dopamine levels in the primate brain. Psychopharmacology 146(2):220–227CrossRefPubMed Chou YH, Karlsson P, Halldin C, Olsson H, Farde L (1999) A PET study of D(1)-like dopamine receptor ligand binding during altered endogenous dopamine levels in the primate brain. Psychopharmacology 146(2):220–227CrossRefPubMed
13.
go back to reference Rinne JO, Laihinen A, Någren K, Bergman J, Haaparanta M, Solin O, Ruotsalainen U, Rinne UK (1991) Positron emission tomography of brain dopamine D-1 receptors with 11C-SCH 23390 in Parkinson’s disease. Acta Radiol Suppl 376:152PubMed Rinne JO, Laihinen A, Någren K, Bergman J, Haaparanta M, Solin O, Ruotsalainen U, Rinne UK (1991) Positron emission tomography of brain dopamine D-1 receptors with 11C-SCH 23390 in Parkinson’s disease. Acta Radiol Suppl 376:152PubMed
14.
go back to reference Ouchi Y, Kanno T, Okada H, Yoshikawa E, Futatsubashi M, Nobezawa S, Torizuka T, Sakamoto M (1999) Presynaptic and postsynaptic dopaminergic binding densities in the nigrostriatal and mesocortical systems in early Parkinson’s disease: a double-tracer positron emission tomography study. Ann Neurol 46(5):723–731CrossRefPubMed Ouchi Y, Kanno T, Okada H, Yoshikawa E, Futatsubashi M, Nobezawa S, Torizuka T, Sakamoto M (1999) Presynaptic and postsynaptic dopaminergic binding densities in the nigrostriatal and mesocortical systems in early Parkinson’s disease: a double-tracer positron emission tomography study. Ann Neurol 46(5):723–731CrossRefPubMed
15.
go back to reference Hurley MJ, Mash DC, Jenner P (2001) Dopamine D(1) receptor expression in human basal ganglia and changes in Parkinson’s disease. Brain Res Mol Brain Res 87(2):271–279CrossRefPubMed Hurley MJ, Mash DC, Jenner P (2001) Dopamine D(1) receptor expression in human basal ganglia and changes in Parkinson’s disease. Brain Res Mol Brain Res 87(2):271–279CrossRefPubMed
16.
go back to reference Shinotoh H, Inoue O, Hirayama K, Aotsuka A, Asahina M, Suhara T, Yamazaki T, Tateno Y (1993) Dopamine D1 receptors in Parkinson’s disease and striatonigral degeneration: a positron emission tomography study. J Neurol Neurosurg Psychiatry 56(5):467–472PubMedCentralCrossRefPubMed Shinotoh H, Inoue O, Hirayama K, Aotsuka A, Asahina M, Suhara T, Yamazaki T, Tateno Y (1993) Dopamine D1 receptors in Parkinson’s disease and striatonigral degeneration: a positron emission tomography study. J Neurol Neurosurg Psychiatry 56(5):467–472PubMedCentralCrossRefPubMed
17.
go back to reference Turjanski N, Lees AJ, Brooks DJ (1997) In vivo studies on striatal dopamine D1 and D2 site binding in L-DOPA treated Parkinson’s disease patients with and without dyskinesias. Neurology 49:717–723CrossRefPubMed Turjanski N, Lees AJ, Brooks DJ (1997) In vivo studies on striatal dopamine D1 and D2 site binding in L-DOPA treated Parkinson’s disease patients with and without dyskinesias. Neurology 49:717–723CrossRefPubMed
18.
go back to reference Cropley VL, Fujita M, Bara-Jimenez W, Brown AK, Zhang XY, Sangare J, Herscovitch P, Pike VW, Hallett M, Nathan PJ, Innis RB (2008) Pre- and post-synaptic dopamine imaging and its relation with frontostriatal cognitive function in Parkinson disease: PET studies with [11C]NNC 112 and [18F]FDOPA. Psychiatry Res 163(2):171–182CrossRefPubMed Cropley VL, Fujita M, Bara-Jimenez W, Brown AK, Zhang XY, Sangare J, Herscovitch P, Pike VW, Hallett M, Nathan PJ, Innis RB (2008) Pre- and post-synaptic dopamine imaging and its relation with frontostriatal cognitive function in Parkinson disease: PET studies with [11C]NNC 112 and [18F]FDOPA. Psychiatry Res 163(2):171–182CrossRefPubMed
19.
go back to reference Laihinen AO, Rinne JO, Ruottinen HM, Någren KA, Lehikoinen PK, Oikonen VJ, Ruotsalainen UH, Rinne UK (1994) PET studies on dopamine D1 receptors in the human brain with carbon-11-SCH 39166 and carbon-11-NNC 756. J Nucl Med 35(12):1916–1920PubMed Laihinen AO, Rinne JO, Ruottinen HM, Någren KA, Lehikoinen PK, Oikonen VJ, Ruotsalainen UH, Rinne UK (1994) PET studies on dopamine D1 receptors in the human brain with carbon-11-SCH 39166 and carbon-11-NNC 756. J Nucl Med 35(12):1916–1920PubMed
20.
go back to reference Ekelund J, Slifstein M, Narendran R, Guillin O, Belani H, Guo NN, Hwang Y, Hwang DR, Abi-Dargham A, Laruelle M (2007) In vivo DA D(1) receptor selectivity of NNC 112 and SCH 23390. Mol Imaging Biol 9(3):117–125CrossRefPubMed Ekelund J, Slifstein M, Narendran R, Guillin O, Belani H, Guo NN, Hwang Y, Hwang DR, Abi-Dargham A, Laruelle M (2007) In vivo DA D(1) receptor selectivity of NNC 112 and SCH 23390. Mol Imaging Biol 9(3):117–125CrossRefPubMed
21.
go back to reference Catafau AM, Searle GE, Bullich S, Gunn RN, Rabiner EA, Herance R, Radua J, Farre M, Laruelle M (2010) Imaging cortical dopamine D1 receptors using [11C]NNC112 and ketanserin blockade of the 5-HT 2A receptors. J Cereb Blood Flow Metab 30(5):985–993PubMedCentralCrossRefPubMed Catafau AM, Searle GE, Bullich S, Gunn RN, Rabiner EA, Herance R, Radua J, Farre M, Laruelle M (2010) Imaging cortical dopamine D1 receptors using [11C]NNC112 and ketanserin blockade of the 5-HT 2A receptors. J Cereb Blood Flow Metab 30(5):985–993PubMedCentralCrossRefPubMed
22.
go back to reference Farde L, Ehrin E, Eriksson L, Greitz T, Hall H, Hedström CG, Litton JE, Sedvall G (1985) Substituted benzamides as ligands for visualization of dopamine receptor binding in the human brain by positron emission tomography. Proc Natl Acad Sci USA 82(11):3863–3867PubMedCentralCrossRefPubMed Farde L, Ehrin E, Eriksson L, Greitz T, Hall H, Hedström CG, Litton JE, Sedvall G (1985) Substituted benzamides as ligands for visualization of dopamine receptor binding in the human brain by positron emission tomography. Proc Natl Acad Sci USA 82(11):3863–3867PubMedCentralCrossRefPubMed
23.
go back to reference Suzuki K, Inoue O, Tamate K, Mikado F (1990) Production of 3-N-[11C]methylspiperone with high specific activity and high radiochemical purity for PET studies: suppression of its radiolysis. Int J Rad Appl Instrum A 41(6):593–599CrossRefPubMed Suzuki K, Inoue O, Tamate K, Mikado F (1990) Production of 3-N-[11C]methylspiperone with high specific activity and high radiochemical purity for PET studies: suppression of its radiolysis. Int J Rad Appl Instrum A 41(6):593–599CrossRefPubMed
24.
go back to reference Mukherjee J, Yang ZY, Brown T, Roemer J, Cooper M (1996) 18F-desmethoxyfallypride: a fluorine-18 labeled radiotracer with properties similar to carbon-11 raclopride for PET imaging studies of dopamine D2 receptors. Life Sci 59(8):669–678CrossRefPubMed Mukherjee J, Yang ZY, Brown T, Roemer J, Cooper M (1996) 18F-desmethoxyfallypride: a fluorine-18 labeled radiotracer with properties similar to carbon-11 raclopride for PET imaging studies of dopamine D2 receptors. Life Sci 59(8):669–678CrossRefPubMed
25.
go back to reference Stark D, Piel M, Hübner H, Gmeiner P, Gründer G, Rösch F (2007) In vitro affinities of various halogenated benzamide derivatives as potential radioligands for non-invasive quantification of D(2)-like dopamine receptors. Bioorg Med Chem 15(21):6819–6829CrossRefPubMed Stark D, Piel M, Hübner H, Gmeiner P, Gründer G, Rösch F (2007) In vitro affinities of various halogenated benzamide derivatives as potential radioligands for non-invasive quantification of D(2)-like dopamine receptors. Bioorg Med Chem 15(21):6819–6829CrossRefPubMed
26.
go back to reference Hwang DR, Kegeles LS, Laruelle M (2000) (-)-N-[(11)C]propyl-norapomorphine: a positron-labeled dopamine agonist for PET imaging of D(2) receptors. Nucl Med Biol 27(6):533–539CrossRefPubMed Hwang DR, Kegeles LS, Laruelle M (2000) (-)-N-[(11)C]propyl-norapomorphine: a positron-labeled dopamine agonist for PET imaging of D(2) receptors. Nucl Med Biol 27(6):533–539CrossRefPubMed
27.
go back to reference Finnema SJ, Seneca N, Farde L, Shchukin E, Sóvágó J, Gulyás B, Wikström HV, Innis RB, Neumeyer JL, Halldin C (2005) A preliminary PET evaluation of the new dopamine D2 receptor agonist [11C]MNPA in cynomolgus monkey. Nucl Med Biol 32(4):353–360CrossRefPubMed Finnema SJ, Seneca N, Farde L, Shchukin E, Sóvágó J, Gulyás B, Wikström HV, Innis RB, Neumeyer JL, Halldin C (2005) A preliminary PET evaluation of the new dopamine D2 receptor agonist [11C]MNPA in cynomolgus monkey. Nucl Med Biol 32(4):353–360CrossRefPubMed
28.
go back to reference Laruelle M (2000) Imaging synaptic neurotransmission with in vivo binding competition techniques: a critical review. J Cereb Blood Flow Metab 20:423–451CrossRefPubMed Laruelle M (2000) Imaging synaptic neurotransmission with in vivo binding competition techniques: a critical review. J Cereb Blood Flow Metab 20:423–451CrossRefPubMed
29.
go back to reference Dentresangle C, Veyre L, le Bars Pierre C, Lavenne F, Pollak P, Guerin J, Froment JC, Brousolle E (1999) Striatal D2 dopamine receptor status in Parkinson’s disease: an 18F DOPA and 11C-raclopride PET study. Mov Disord 14(6):1025–1030CrossRefPubMed Dentresangle C, Veyre L, le Bars Pierre C, Lavenne F, Pollak P, Guerin J, Froment JC, Brousolle E (1999) Striatal D2 dopamine receptor status in Parkinson’s disease: an 18F DOPA and 11C-raclopride PET study. Mov Disord 14(6):1025–1030CrossRefPubMed
30.
go back to reference Kaasinen V, Ruottinen HM, Nagren K, Lehikoinen P, Oikonen V, Rinne JO (2000) Upregulation of putaminal dopamine D2 receptors in early Parkinson’s disease: a comparative PET study with 11C-raclopride and 11C-methylspiperone. J Nucl Med 41:65–70PubMed Kaasinen V, Ruottinen HM, Nagren K, Lehikoinen P, Oikonen V, Rinne JO (2000) Upregulation of putaminal dopamine D2 receptors in early Parkinson’s disease: a comparative PET study with 11C-raclopride and 11C-methylspiperone. J Nucl Med 41:65–70PubMed
31.
go back to reference Sawle GV, Playford ED, Brooks DJ, Quinn N, Frackowiak RS (1993) Asymmetrical presynaptic and postsynaptic changes in the striatal dopamine projection in Dopa naïve parkinsonism. Diagnostic implications of the D2 receptor status. Brain 116:853–867CrossRefPubMed Sawle GV, Playford ED, Brooks DJ, Quinn N, Frackowiak RS (1993) Asymmetrical presynaptic and postsynaptic changes in the striatal dopamine projection in Dopa naïve parkinsonism. Diagnostic implications of the D2 receptor status. Brain 116:853–867CrossRefPubMed
32.
go back to reference Antonini A, Schwarz J, Oertel WH, Pogarell O, Leenders KL (1997) 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 12(1):33–38CrossRefPubMed Antonini A, Schwarz J, Oertel WH, Pogarell O, Leenders KL (1997) 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 12(1):33–38CrossRefPubMed
33.
go back to reference Rinne JO, Laihinen A, Någren K, Bergman J, Solin O, Haaparanta M, Ruotsalainen U, Rinne UK (1990) PET demonstrates different behaviour of striatal dopamine D-1 and D-2 receptors in early Parkinson’s disease. J Neurosci Res 27(4):494–499CrossRefPubMed Rinne JO, Laihinen A, Någren K, Bergman J, Solin O, Haaparanta M, Ruotsalainen U, Rinne UK (1990) PET demonstrates different behaviour of striatal dopamine D-1 and D-2 receptors in early Parkinson’s disease. J Neurosci Res 27(4):494–499CrossRefPubMed
34.
go back to reference Rinne JO, Laihinen A, Rinne UK, Någren K, Bergman J, Ruotsalainen U (1993) PET study on striatal dopamine D2 receptor changes during the progression of early Parkinson’s disease. Mov Disord 8(2):134–138CrossRefPubMed Rinne JO, Laihinen A, Rinne UK, Någren K, Bergman J, Ruotsalainen U (1993) PET study on striatal dopamine D2 receptor changes during the progression of early Parkinson’s disease. Mov Disord 8(2):134–138CrossRefPubMed
35.
go back to reference Rinne JO, Laihinen A, Ruottinen H, Ruotsalainen U, Någren K, Lehikoinen P, Oikonen V, Rinne UK (1995) Increased density of dopamine D2 receptors in the putamen, but not in the caudate nucleus in early Parkinson’s disease: a PET study with [11C]raclopride. J Neurol Sci 132(2):156–161CrossRefPubMed Rinne JO, Laihinen A, Ruottinen H, Ruotsalainen U, Någren K, Lehikoinen P, Oikonen V, Rinne UK (1995) Increased density of dopamine D2 receptors in the putamen, but not in the caudate nucleus in early Parkinson’s disease: a PET study with [11C]raclopride. J Neurol Sci 132(2):156–161CrossRefPubMed
36.
go back to reference Schwarting RK, Huston JP (1996) Unilateral 6 hydroxy dopamine lesions in meso striatal dopamine neurons and their physiological sequelae. Prog Neurobiol 49:215–266CrossRefPubMed Schwarting RK, Huston JP (1996) Unilateral 6 hydroxy dopamine lesions in meso striatal dopamine neurons and their physiological sequelae. Prog Neurobiol 49:215–266CrossRefPubMed
37.
go back to reference Brooks DJ, Ibanez V, Saule GV, Playford ED, Quinn N, Mathias CJ, Lees AJ, Marsden CD, Bannister R, Frackowiak RS (1992) 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 31:184–192CrossRefPubMed Brooks DJ, Ibanez V, Saule GV, Playford ED, Quinn N, Mathias CJ, Lees AJ, Marsden CD, Bannister R, Frackowiak RS (1992) 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 31:184–192CrossRefPubMed
38.
go back to reference Antonini A, Schwarz J, Oertel WH, Beer HF, Madeja UD, Leenders KL (1994) [11C]raclopride and positron emission tomography in previously untreated patients with Parkinson’s disease: influence of L-dopa and lisuride therapy on striatal dopamine D2-receptors. Neurology 44(7):1325–1329CrossRefPubMed Antonini A, Schwarz J, Oertel WH, Beer HF, Madeja UD, Leenders KL (1994) [11C]raclopride and positron emission tomography in previously untreated patients with Parkinson’s disease: influence of L-dopa and lisuride therapy on striatal dopamine D2-receptors. Neurology 44(7):1325–1329CrossRefPubMed
39.
go back to reference Thobois S, Vingerhoets F, Fraix V, Xie-Brustolin J, Mollion H, Costes N, Mertens P, Benebid AL, Pollak P, Broussolle E (2004) Role of dopaminergic treatment in dopamine receptor downregulation in advanced Parkinson’s disease: a positron emission tomographic study. Arch Neurol 61(11):1705–2179CrossRefPubMed Thobois S, Vingerhoets F, Fraix V, Xie-Brustolin J, Mollion H, Costes N, Mertens P, Benebid AL, Pollak P, Broussolle E (2004) Role of dopaminergic treatment in dopamine receptor downregulation in advanced Parkinson’s disease: a positron emission tomographic study. Arch Neurol 61(11):1705–2179CrossRefPubMed
40.
go back to reference Kempster PA, Gibb WRG, Stern GM, Lees AJ (1989) Asymmetry of substantia nigra neuronal loss in Parkinson’s disease and its relevance to the mechanism of levodopa related motor fluctuations. J Neurol Neurosurg Psychiatry 52:72–76PubMedCentralCrossRefPubMed Kempster PA, Gibb WRG, Stern GM, Lees AJ (1989) Asymmetry of substantia nigra neuronal loss in Parkinson’s disease and its relevance to the mechanism of levodopa related motor fluctuations. J Neurol Neurosurg Psychiatry 52:72–76PubMedCentralCrossRefPubMed
41.
go back to reference de la Fuente Fernandez R, Pal PK, Vingerhoets FJG, Kishore A, Schulzer M, Mak EK, Ruth TJ, Snow BJ, Calne DB, Stoessl AJ (2000) Evidence for impaired presynaptic dopamine function in Parkinsonian patients with motor fluctuations. J Neural Trasm 107:49–57CrossRef de la Fuente Fernandez R, Pal PK, Vingerhoets FJG, Kishore A, Schulzer M, Mak EK, Ruth TJ, Snow BJ, Calne DB, Stoessl AJ (2000) Evidence for impaired presynaptic dopamine function in Parkinsonian patients with motor fluctuations. J Neural Trasm 107:49–57CrossRef
42.
go back to reference Landwehrmeyer B, Mengod G, Palacios JM (1993) Dopamine D3 receptor mRNA and binding sites in human brain. Brain Res Mol Brain Res 18:187–192CrossRefPubMed Landwehrmeyer B, Mengod G, Palacios JM (1993) Dopamine D3 receptor mRNA and binding sites in human brain. Brain Res Mol Brain Res 18:187–192CrossRefPubMed
43.
go back to reference Murray AM, Ryoo HL, Gurevich E, Joyce JN (1994) Localization of dopamine D3 receptors to mesolimbic and D2 receptors to mesostriatal regions of human forebrain. Proc Natl Acad Sci USA 91:11271–11275PubMedCentralCrossRefPubMed Murray AM, Ryoo HL, Gurevich E, Joyce JN (1994) Localization of dopamine D3 receptors to mesolimbic and D2 receptors to mesostriatal regions of human forebrain. Proc Natl Acad Sci USA 91:11271–11275PubMedCentralCrossRefPubMed
44.
go back to reference Wilson AA, McCormick P, Kapur S, Willeit M, Garcia A, Hussey D, Houle S, Seeman P, Ginovart N (2005) Radiosynthesis and evaluation of [11C]-(+)-4-propyl-3,4,4a,5,6, 10b-hexahydro-2H-naphtho[1,2-b][1,4]oxazin-9 -ol as a potential radiotracer for in vivo imaging of the dopamine D2 high-affinity state with positron emission tomography. J Med Chem 48:4153CrossRefPubMed Wilson AA, McCormick P, Kapur S, Willeit M, Garcia A, Hussey D, Houle S, Seeman P, Ginovart N (2005) Radiosynthesis and evaluation of [11C]-(+)-4-propyl-3,4,4a,5,6, 10b-hexahydro-2H-naphtho[1,2-b][1,4]oxazin-9 -ol as a potential radiotracer for in vivo imaging of the dopamine D2 high-affinity state with positron emission tomography. J Med Chem 48:4153CrossRefPubMed
45.
go back to reference Seeman P, Ulpian C, Larsen RD, Anderson PS (1993) Dopamine receptors labelled by PHNO. Synapse 14:254–262CrossRefPubMed Seeman P, Ulpian C, Larsen RD, Anderson PS (1993) Dopamine receptors labelled by PHNO. Synapse 14:254–262CrossRefPubMed
46.
go back to reference Willeit M, Ginovart N, Kapur S, Houle S, Hussey D, Seeman P, Wilson AA (2006) High-affinity states of human brain dopamine D2/3 receptors imaged by the agonist [11C]-(+)-PHNO. Biol Psychiatry 59:389–394CrossRefPubMed Willeit M, Ginovart N, Kapur S, Houle S, Hussey D, Seeman P, Wilson AA (2006) High-affinity states of human brain dopamine D2/3 receptors imaged by the agonist [11C]-(+)-PHNO. Biol Psychiatry 59:389–394CrossRefPubMed
47.
go back to reference Graff-Guerrero A, Willeit M, Ginovart N, Mamo D, Mizrahi R, Rusjan P, Vitcu I, Seeman P, Wilson AA, Kapur S (2008) Brain region binding of the D(2/3) agonist [(11)C]-(+)-PHNO and the D(2/3) antagonist [(11)C]raclopride in healthy humans. Hum Brain Mapp 29:400–410CrossRefPubMed Graff-Guerrero A, Willeit M, Ginovart N, Mamo D, Mizrahi R, Rusjan P, Vitcu I, Seeman P, Wilson AA, Kapur S (2008) Brain region binding of the D(2/3) agonist [(11)C]-(+)-PHNO and the D(2/3) antagonist [(11)C]raclopride in healthy humans. Hum Brain Mapp 29:400–410CrossRefPubMed
48.
go back to reference Rabiner EA, Slifstein M, Nobrega J, Plisson C, Huiban M, Raymond R, Diwan M, Wilson AA, McCormick P, Gentile G, Gunn RN, Laruelle MA (2009) In vivo quantification of regional dopamine-D3 receptor binding potential of (+)-PHNO: studies in non-human primates and transgenic mice. Synapse 63(9):782–793CrossRefPubMed Rabiner EA, Slifstein M, Nobrega J, Plisson C, Huiban M, Raymond R, Diwan M, Wilson AA, McCormick P, Gentile G, Gunn RN, Laruelle MA (2009) In vivo quantification of regional dopamine-D3 receptor binding potential of (+)-PHNO: studies in non-human primates and transgenic mice. Synapse 63(9):782–793CrossRefPubMed
49.
go back to reference Boileau I, Guttman M, Rusjan P, Adams JR, Houle S, Tong J, Hornykiewicz O, Furukawa Y, Wilson AA, Kapur S, Kish SJ (2009) Decreased binding of the D3 dopamine receptor-preferring ligand [11C]-(+)-PHNO in drug-naive Parkinson’s disease. Brain 132(Pt 5):1366–1375CrossRefPubMed Boileau I, Guttman M, Rusjan P, Adams JR, Houle S, Tong J, Hornykiewicz O, Furukawa Y, Wilson AA, Kapur S, Kish SJ (2009) Decreased binding of the D3 dopamine receptor-preferring ligand [11C]-(+)-PHNO in drug-naive Parkinson’s disease. Brain 132(Pt 5):1366–1375CrossRefPubMed
50.
go back to reference Hattori N, Kitada T, Matsumine H, Asakawa S, Yamamura Y, Yoshino H, Kobayashi T, Yokochi M, Wang M, Yoritaka A, Kondo T, Kuzuhara S, Nakamura S, Shimizu N, Mizuno Y (1998) Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann Neurol 44:935–941CrossRefPubMed Hattori N, Kitada T, Matsumine H, Asakawa S, Yamamura Y, Yoshino H, Kobayashi T, Yokochi M, Wang M, Yoritaka A, Kondo T, Kuzuhara S, Nakamura S, Shimizu N, Mizuno Y (1998) Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann Neurol 44:935–941CrossRefPubMed
51.
go back to reference Abbas N, Lücking CB, Ricard S, Dürr A, Bonifati V, De Michele G, Bouley S, Vaughan JR, Gasser T, Marconi R, Broussolle E, Brefel-Courbon C, Harhangi BS, Oostra BA, Fabrizio E, Böhme GA, Pradier L, Wood NW, Filla A, Meco G, Denefle P, Agid Y, Brice A (1999) A wide variety of mutations in the parkin gene are responsible for autosomal recessive parkinsonism in Europe. French Parkinson’s Disease Genetics Study Group and the European Consortium on Genetic Susceptibility in Parkinson’s disease. Hum Mol Genet 8(4):567–574CrossRefPubMed Abbas N, Lücking CB, Ricard S, Dürr A, Bonifati V, De Michele G, Bouley S, Vaughan JR, Gasser T, Marconi R, Broussolle E, Brefel-Courbon C, Harhangi BS, Oostra BA, Fabrizio E, Böhme GA, Pradier L, Wood NW, Filla A, Meco G, Denefle P, Agid Y, Brice A (1999) A wide variety of mutations in the parkin gene are responsible for autosomal recessive parkinsonism in Europe. French Parkinson’s Disease Genetics Study Group and the European Consortium on Genetic Susceptibility in Parkinson’s disease. Hum Mol Genet 8(4):567–574CrossRefPubMed
52.
go back to reference Shimura H, Hattori N, Si Kubo, Mizuno Y, Asakawa S, Minoshima S, Shimizu N, Iwai K, Chiba T, Tanaka K, Suzuki T (2000) Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet 25(3):302–305CrossRefPubMed Shimura H, Hattori N, Si Kubo, Mizuno Y, Asakawa S, Minoshima S, Shimizu N, Iwai K, Chiba T, Tanaka K, Suzuki T (2000) Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet 25(3):302–305CrossRefPubMed
53.
go back to reference Zimprich A, Biskup S, Leitner P, Lichner M, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti R, Calne D (2004) Mutations in LRRK2 cause autosomal dominant parkinsonism with pleomorphic pathology. Neuron 44(4):601–607CrossRefPubMed Zimprich A, Biskup S, Leitner P, Lichner M, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti R, Calne D (2004) Mutations in LRRK2 cause autosomal dominant parkinsonism with pleomorphic pathology. Neuron 44(4):601–607CrossRefPubMed
54.
go back to reference Scherfler C, Khan NL, Pavese N, Eunson L, Graham E, Lees AJ, Quinn NP, Wood NW, Brooks DJ, Piccini P (2004) Striatal and cortical pre and postsynaptic dopaminergic dysfunction in sporadic parkin linked parkinsonism. Brain 127(6):1332–1342CrossRefPubMed Scherfler C, Khan NL, Pavese N, Eunson L, Graham E, Lees AJ, Quinn NP, Wood NW, Brooks DJ, Piccini P (2004) Striatal and cortical pre and postsynaptic dopaminergic dysfunction in sporadic parkin linked parkinsonism. Brain 127(6):1332–1342CrossRefPubMed
55.
go back to reference Scherfler C, Khan NL, Pavese N, Lees AJ, Quinn NP, Brooks DJ, Piccini P (2006) Upregulation of dopamine D2 receptors in dopaminergic drug naïve patients with parkin gene mutations. Mov Disord 21(6):783–788CrossRefPubMed Scherfler C, Khan NL, Pavese N, Lees AJ, Quinn NP, Brooks DJ, Piccini P (2006) Upregulation of dopamine D2 receptors in dopaminergic drug naïve patients with parkin gene mutations. Mov Disord 21(6):783–788CrossRefPubMed
56.
go back to reference Endres CJ, Kolachana BS, Saunders RC, Su T, Weinberger D, Breier A, Eckelman WC, Carson RE (1997) Kinetic modeling of [11C]raclopride: combined PET-microdialysis studies. Cereb Blood Flow Metab 17(9):932–942CrossRef Endres CJ, Kolachana BS, Saunders RC, Su T, Weinberger D, Breier A, Eckelman WC, Carson RE (1997) Kinetic modeling of [11C]raclopride: combined PET-microdialysis studies. Cereb Blood Flow Metab 17(9):932–942CrossRef
57.
go back to reference Litvan I (1999) Recent advances in atypical parkinsonian disorders. Curr Opin Neurol 12(4):441–446CrossRefPubMed Litvan I (1999) Recent advances in atypical parkinsonian disorders. Curr Opin Neurol 12(4):441–446CrossRefPubMed
58.
go back to reference Schreckenberger M, Hägele S, Siessmeier T, Buchholz HG, Armbrust-Henrich H, Rösch F, Gründer G, Bartenstein P, Vogt T (2004) The dopamine D2 receptor ligand 18F-desmethoxyfallypride: an appropriate fluorinated PET tracer for the differential diagnosis of parkinsonism. Eur J Nucl Med Mol Imaging 31(8):1128–1135CrossRefPubMed Schreckenberger M, Hägele S, Siessmeier T, Buchholz HG, Armbrust-Henrich H, Rösch F, Gründer G, Bartenstein P, Vogt T (2004) The dopamine D2 receptor ligand 18F-desmethoxyfallypride: an appropriate fluorinated PET tracer for the differential diagnosis of parkinsonism. Eur J Nucl Med Mol Imaging 31(8):1128–1135CrossRefPubMed
59.
go back to reference la Fougère C, Pöpperl G, Levin J, Wängler B, Böning G, Uebleis C, Cumming P, Bartenstein P, Bötzel K, Tatsch K (2010) The value of the dopamine D2/3 receptor ligand 18F-desmethoxyfallypride for the differentiation of idiopathic and nonidiopathic parkinsonian syndromes. J Nucl Med 51(4):581–587CrossRefPubMed la Fougère C, Pöpperl G, Levin J, Wängler B, Böning G, Uebleis C, Cumming P, Bartenstein P, Bötzel K, Tatsch K (2010) The value of the dopamine D2/3 receptor ligand 18F-desmethoxyfallypride for the differentiation of idiopathic and nonidiopathic parkinsonian syndromes. J Nucl Med 51(4):581–587CrossRefPubMed
60.
go back to reference Hellwig S, Amtage F, Kreft A, Buchert R, Winz OH, Vach W, Spehl TS, Rijntjes M, Hellwig B, Weiller C, Winkler C, Weber WA, Tüscher O, Meyer PT (2012) [18F]FDG-PET is superior to [123I]IBZM-SPECT for the differential diagnosis of parkinsonism. Neurology 79(13):1314–1322CrossRefPubMed Hellwig S, Amtage F, Kreft A, Buchert R, Winz OH, Vach W, Spehl TS, Rijntjes M, Hellwig B, Weiller C, Winkler C, Weber WA, Tüscher O, Meyer PT (2012) [18F]FDG-PET is superior to [123I]IBZM-SPECT for the differential diagnosis of parkinsonism. Neurology 79(13):1314–1322CrossRefPubMed
61.
go back to reference Politis M, Piccini P, Pavese N, Koh SB, Brooks DJ (2008) Evidence of dopamine dysfunction in the hypothalamus of patients with Parkinson’s disease: an in vivo 11C-raclopride PET study. Exp Neurol 214:112–116CrossRefPubMed Politis M, Piccini P, Pavese N, Koh SB, Brooks DJ (2008) Evidence of dopamine dysfunction in the hypothalamus of patients with Parkinson’s disease: an in vivo 11C-raclopride PET study. Exp Neurol 214:112–116CrossRefPubMed
62.
go back to reference Halldin C, Farde L, Högberg T, Mohell N, Hall H, Suhara T, Karlsson P, Nakashima Y, Swahn CG (1995) Carbon-11-FLB 457: a radioligand for extrastriatal D2 dopamine receptors. J Nucl Med 36(7):1275–1281PubMed Halldin C, Farde L, Högberg T, Mohell N, Hall H, Suhara T, Karlsson P, Nakashima Y, Swahn CG (1995) Carbon-11-FLB 457: a radioligand for extrastriatal D2 dopamine receptors. J Nucl Med 36(7):1275–1281PubMed
63.
go back to reference Kaasinen V, Någren K, Hietala J, Oikonen V, Vilkman H, Farde L, Halldin C, Rinne JO (2000) Extrastriatal dopamine D2 and D3 receptors in early and advanced Parkinson’s disease. Neurology 54(7):1482–1487CrossRefPubMed Kaasinen V, Någren K, Hietala J, Oikonen V, Vilkman H, Farde L, Halldin C, Rinne JO (2000) Extrastriatal dopamine D2 and D3 receptors in early and advanced Parkinson’s disease. Neurology 54(7):1482–1487CrossRefPubMed
64.
go back to reference Kaasinen V, Aalto S, Nagren K, Hietala J, Sonninen P, Rinne JO (2003) Extrastriatal dopamine D(2) receptors in Parkinson’s disease: a longitudinal study. J Neural Transm 110(6):591–601CrossRefPubMed Kaasinen V, Aalto S, Nagren K, Hietala J, Sonninen P, Rinne JO (2003) Extrastriatal dopamine D(2) receptors in Parkinson’s disease: a longitudinal study. J Neural Transm 110(6):591–601CrossRefPubMed
65.
go back to reference Doudet DJ, Holden JE (2003) Raclopride studies of dopamine release: dependence on presynaptic integrity. Biol Psychiatry 54:1193–1199CrossRefPubMed Doudet DJ, Holden JE (2003) Raclopride studies of dopamine release: dependence on presynaptic integrity. Biol Psychiatry 54:1193–1199CrossRefPubMed
66.
go back to reference Breier A, Su TP, Saunders R, Carson RE, Kolachana BS, de Bartolomeis A, Weinberger DR, Weisenfeld N, Malhotra AK, Eckelman WC, Pickar D (1997) Schizophrenia is associated with elevated amphetamine-induced synaptic dopamine concentrations: evidence from a novel positron emission tomography method. Proc Natl Acad Sci USA 94(6):2569–2574PubMedCentralCrossRefPubMed Breier A, Su TP, Saunders R, Carson RE, Kolachana BS, de Bartolomeis A, Weinberger DR, Weisenfeld N, Malhotra AK, Eckelman WC, Pickar D (1997) Schizophrenia is associated with elevated amphetamine-induced synaptic dopamine concentrations: evidence from a novel positron emission tomography method. Proc Natl Acad Sci USA 94(6):2569–2574PubMedCentralCrossRefPubMed
67.
go back to reference Goerendt IK, Messa C, Lawrence AD, Grasby PM, Piccini P, Brooks DJ (2003) Dopamine release during sequential finger movements in health and Parkinson’s disease: a PET study. Brain 126:312–325CrossRefPubMed Goerendt IK, Messa C, Lawrence AD, Grasby PM, Piccini P, Brooks DJ (2003) Dopamine release during sequential finger movements in health and Parkinson’s disease: a PET study. Brain 126:312–325CrossRefPubMed
68.
go back to reference Tedroff J, Pederson M, Aquilonius SM, Hartvig P, Jacobsson G, Langstrom B (1996) Levodopa induced changes in synaptic dopamine in patients with Parkinson’s disease as measured by 11C-raclopride displacement and PET. Neurology 46:1430–1436CrossRefPubMed Tedroff J, Pederson M, Aquilonius SM, Hartvig P, Jacobsson G, Langstrom B (1996) Levodopa induced changes in synaptic dopamine in patients with Parkinson’s disease as measured by 11C-raclopride displacement and PET. Neurology 46:1430–1436CrossRefPubMed
69.
go back to reference Piccini P, Pavese N, Brooks DJ (2003) Endogenous dopamine release after pharmacological challenges in Parkinson’s disease. Ann Neurol 53:647–653CrossRefPubMed Piccini P, Pavese N, Brooks DJ (2003) Endogenous dopamine release after pharmacological challenges in Parkinson’s disease. Ann Neurol 53:647–653CrossRefPubMed
70.
go back to reference Koochesfahani KM, de la Fuerte-Fernandes R, Sossi V, Schulzer M, Lakshmi N, Yatham LN, Ruth TJ, Blinder S, Stoessl AJ (2006) Oral methylphenidate fails to elicit significant changes in extracellular putaminal dopamine levels in Parkinson’s disease patients: PET studies. Mov Disord 21(7):970–975CrossRefPubMed Koochesfahani KM, de la Fuerte-Fernandes R, Sossi V, Schulzer M, Lakshmi N, Yatham LN, Ruth TJ, Blinder S, Stoessl AJ (2006) Oral methylphenidate fails to elicit significant changes in extracellular putaminal dopamine levels in Parkinson’s disease patients: PET studies. Mov Disord 21(7):970–975CrossRefPubMed
71.
go back to reference Sawamoto N, Piccini P, Hotten G, Pavese N, Thielemans K, Brooks DJ (2008) Cognitive deficits and striatal-frontal dopamine release in Parkinson’s disease. Brain 131:1294–1302CrossRefPubMed Sawamoto N, Piccini P, Hotten G, Pavese N, Thielemans K, Brooks DJ (2008) Cognitive deficits and striatal-frontal dopamine release in Parkinson’s disease. Brain 131:1294–1302CrossRefPubMed
72.
go back to reference de la Fuente-Fernandez R, Lu JQ, Sossi V, Jivan S, Schulzer M, Holden JE, Lee CS, Ruth TJ, Calne DB, Stoessl AJ (2001) Biochemical variations in the synaptic levels of dopamine precede motor fluctuations in Parkinson’s disease: PET evidence of increased dopamine turnover. Ann Neurol 49:298–303CrossRefPubMed de la Fuente-Fernandez R, Lu JQ, Sossi V, Jivan S, Schulzer M, Holden JE, Lee CS, Ruth TJ, Calne DB, Stoessl AJ (2001) Biochemical variations in the synaptic levels of dopamine precede motor fluctuations in Parkinson’s disease: PET evidence of increased dopamine turnover. Ann Neurol 49:298–303CrossRefPubMed
73.
go back to reference de la Fuente-Fernández R, Lim AS, Sossi V, Holden JE, Calne DB, Ruth TJ, Stoessl AJ (2001) Apomorphine-induced changes in synaptic dopamine levels: positron emission tomography evidence for presynaptic inhibition. J Cereb Blood Flow Metab 21:1151–1159CrossRef de la Fuente-Fernández R, Lim AS, Sossi V, Holden JE, Calne DB, Ruth TJ, Stoessl AJ (2001) Apomorphine-induced changes in synaptic dopamine levels: positron emission tomography evidence for presynaptic inhibition. J Cereb Blood Flow Metab 21:1151–1159CrossRef
74.
go back to reference de la Fuente-Fernandez R, Sossi V, Huang Z, Furtado S, Lu QR, Calne DB, Ruth TJ, Stoessl AJ (2004) Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: implications for dyskinesias. Brain 127:2747–2754CrossRefPubMed de la Fuente-Fernandez R, Sossi V, Huang Z, Furtado S, Lu QR, Calne DB, Ruth TJ, Stoessl AJ (2004) Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: implications for dyskinesias. Brain 127:2747–2754CrossRefPubMed
75.
go back to reference Pavese N, Evans AH, Tai YF, Hotton G, Brooks DJ, Lees AJ, Piccini P (2006) Clinical correlates of levodopa induced dopamine release in Parkinson’s disease: a PET study. Neurology 67:1612–1617CrossRefPubMed Pavese N, Evans AH, Tai YF, Hotton G, Brooks DJ, Lees AJ, Piccini P (2006) Clinical correlates of levodopa induced dopamine release in Parkinson’s disease: a PET study. Neurology 67:1612–1617CrossRefPubMed
76.
go back to reference Piccini P, Brooks DJ, Bjorklund A, Gunn RN, Grasby PM, Rimoldi O, Brundin P, Hagell P, Rehncrona S, Widner H, Lindvall O (1999) Dopamine release from nigral transplants visualised in vivo in a Parkinson’s patient. Nat Neurosci 2(12):1047–1048CrossRef Piccini P, Brooks DJ, Bjorklund A, Gunn RN, Grasby PM, Rimoldi O, Brundin P, Hagell P, Rehncrona S, Widner H, Lindvall O (1999) Dopamine release from nigral transplants visualised in vivo in a Parkinson’s patient. Nat Neurosci 2(12):1047–1048CrossRef
77.
go back to reference Piccini P, Pavese N, Hagell P, Reiner J, Bjorklund A, Oertel WH, Quinn NP, Brooks DJ, Lindvall O (2005) Factors affecting the clinical outcome after neural transplantation in Parkinson’s disease. Brain 128(12):2977–2986CrossRefPubMed Piccini P, Pavese N, Hagell P, Reiner J, Bjorklund A, Oertel WH, Quinn NP, Brooks DJ, Lindvall O (2005) Factors affecting the clinical outcome after neural transplantation in Parkinson’s disease. Brain 128(12):2977–2986CrossRefPubMed
78.
go back to reference Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Rehncrona S, Bjorklund A, Lindvall O, Piccini P (2010) Serotonergic neurons mediate dyskinesia side effects in Parkinson’s patients with neural transplants. Sci Transl Med 2(38):38–46CrossRef Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Rehncrona S, Bjorklund A, Lindvall O, Piccini P (2010) Serotonergic neurons mediate dyskinesia side effects in Parkinson’s patients with neural transplants. Sci Transl Med 2(38):38–46CrossRef
79.
go back to reference Politis M, Oertel WH, Wu K, Quinn NP, Pogarell O, Brooks DJ, Bjorklund A, Lindvall O, Piccini P (2011) Graft-induced dyskinesias in Parkinson’s disease: high striatal serotonin/dopamine transporter ratio. Mov Disord 26(11):1997–2003CrossRefPubMed Politis M, Oertel WH, Wu K, Quinn NP, Pogarell O, Brooks DJ, Bjorklund A, Lindvall O, Piccini P (2011) Graft-induced dyskinesias in Parkinson’s disease: high striatal serotonin/dopamine transporter ratio. Mov Disord 26(11):1997–2003CrossRefPubMed
80.
go back to reference Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Oertel WH, Björklund A, Lindvall O, Piccini P (2012) Serotonin neuron loss and nonmotor symptoms continue in Parkinson’s patients treated with dopamine grafts. Sci Transl Med 4(128):128–141CrossRef Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Oertel WH, Björklund A, Lindvall O, Piccini P (2012) Serotonin neuron loss and nonmotor symptoms continue in Parkinson’s patients treated with dopamine grafts. Sci Transl Med 4(128):128–141CrossRef
81.
go back to reference Ma Y, Feigin A, Dhawan V, Fukuda M, Shi Q, Greene P, Breeze R, Fahn S, Freed C, Eidelberg D (2002) Dyskinesia after fetal cell transplantation for Parkinsonism: a PET study. Ann Neurol 52:628–634CrossRefPubMed Ma Y, Feigin A, Dhawan V, Fukuda M, Shi Q, Greene P, Breeze R, Fahn S, Freed C, Eidelberg D (2002) Dyskinesia after fetal cell transplantation for Parkinsonism: a PET study. Ann Neurol 52:628–634CrossRefPubMed
82.
go back to reference Hagell P, Piccini P, Björklund A, Brundin P, Rehncrona S, Widner H, Crabb L, Pavese N, Oertel WH, Quinn N, Brooks DJ, Lindvall O (2002) Dyskinesias following neural transplantation in Parkinson’s disease. Nat Neurosci 5(7):627–628PubMed Hagell P, Piccini P, Björklund A, Brundin P, Rehncrona S, Widner H, Crabb L, Pavese N, Oertel WH, Quinn N, Brooks DJ, Lindvall O (2002) Dyskinesias following neural transplantation in Parkinson’s disease. Nat Neurosci 5(7):627–628PubMed
83.
go back to reference Molina JA, Sáinz-Artiga MJ, Fraile A, Jiménez-Jiménez FJ, Villanueva C, Ortí-Pareja M, Bermejo F (2000) Pathologic gambling in Parkinson’s disease: a behavioral manifestation of pharmacologic treatment? Mov Disord 15(5):869–872CrossRefPubMed Molina JA, Sáinz-Artiga MJ, Fraile A, Jiménez-Jiménez FJ, Villanueva C, Ortí-Pareja M, Bermejo F (2000) Pathologic gambling in Parkinson’s disease: a behavioral manifestation of pharmacologic treatment? Mov Disord 15(5):869–872CrossRefPubMed
84.
go back to reference Voon V, Reynolds B, Brezing C, Galleo C, Skaljic M, Ekanayake V, Fernandez H, Potenza MN, Dolan RJ, Hallett M (2010) Impulsive choice and response in dopamine agonist-related impulse control behaviours. Psychopharmacology 207(4):645–659PubMedCentralCrossRefPubMed Voon V, Reynolds B, Brezing C, Galleo C, Skaljic M, Ekanayake V, Fernandez H, Potenza MN, Dolan RJ, Hallett M (2010) Impulsive choice and response in dopamine agonist-related impulse control behaviours. Psychopharmacology 207(4):645–659PubMedCentralCrossRefPubMed
85.
go back to reference McElroy SL, Hudson JI, Pope HJ, Keck PE, Aizley HG (1992) The DSM-III-R impulse control disorders not elsewhere classified: clinical characteristics and relationships to other psychiatric disorders. Am J Psychiatry 149:318–327CrossRefPubMed McElroy SL, Hudson JI, Pope HJ, Keck PE, Aizley HG (1992) The DSM-III-R impulse control disorders not elsewhere classified: clinical characteristics and relationships to other psychiatric disorders. Am J Psychiatry 149:318–327CrossRefPubMed
86.
go back to reference Giovannoni G, O’Sullivan JD, Turner K, Manson AJ, Lees AJ (2000) Hedonistic homeostasis dysregulation in patients with Parkinson’s disease on dopamine replacement therapies. J Neurol Neurosurg Psychiatry 68:423–428PubMedCentralCrossRefPubMed Giovannoni G, O’Sullivan JD, Turner K, Manson AJ, Lees AJ (2000) Hedonistic homeostasis dysregulation in patients with Parkinson’s disease on dopamine replacement therapies. J Neurol Neurosurg Psychiatry 68:423–428PubMedCentralCrossRefPubMed
87.
go back to reference Evans AH, Pavese N, Lawrence AD, Tai FY, Appel S, Doder M, Brooks DJ, Lees AJ, Piccini P (2006) Compulsive drug use linked to sensitized ventral striatal dopamine transmission. Ann Neurol 59:852–858CrossRefPubMed Evans AH, Pavese N, Lawrence AD, Tai FY, Appel S, Doder M, Brooks DJ, Lees AJ, Piccini P (2006) Compulsive drug use linked to sensitized ventral striatal dopamine transmission. Ann Neurol 59:852–858CrossRefPubMed
88.
go back to reference Steeves TDL, Miyasaki J, Zurowski M, Lang AE, Pellecchia G, van Elmeren T, Rusjan P, Houle S, Strafella AP (2009) Increased striatal dopamine release in Parkinsonian patients with pathological gambling: a 11C-raclopride PET study. Brain 132:1376–1385PubMedCentralCrossRefPubMed Steeves TDL, Miyasaki J, Zurowski M, Lang AE, Pellecchia G, van Elmeren T, Rusjan P, Houle S, Strafella AP (2009) Increased striatal dopamine release in Parkinsonian patients with pathological gambling: a 11C-raclopride PET study. Brain 132:1376–1385PubMedCentralCrossRefPubMed
89.
go back to reference Callesen MB, Hansen KV, Gjedde A, Linnet J, Møller A (2013) Dopaminergic and clinical correlates of pathological gambling in Parkinson’s disease: a case report. Front Behav Neurosci 7:95PubMedCentralCrossRefPubMed Callesen MB, Hansen KV, Gjedde A, Linnet J, Møller A (2013) Dopaminergic and clinical correlates of pathological gambling in Parkinson’s disease: a case report. Front Behav Neurosci 7:95PubMedCentralCrossRefPubMed
90.
go back to reference O’Sullivan SS, Wu K, Politis M, Lawrence AD, Evans AH, Bose SK, Djamshidian A, Lees AJ, Piccini P (2011) Cue-induced striatal dopamine release in Parkinson’s disease-associated impulsive-compulsive behaviours. Brain 134(Pt 4):969–978CrossRefPubMed O’Sullivan SS, Wu K, Politis M, Lawrence AD, Evans AH, Bose SK, Djamshidian A, Lees AJ, Piccini P (2011) Cue-induced striatal dopamine release in Parkinson’s disease-associated impulsive-compulsive behaviours. Brain 134(Pt 4):969–978CrossRefPubMed
91.
go back to reference Politis M, Loane C, Wu K, O’Sullivan SS, Woodhead Z, Kiferle L, Lawrence AD, Lees AJ, Piccini P (2013) Neural response to visual sexual cues in dopamine treatment-linked hypersexuality in Parkinson’s disease. Brain 136(Pt 2):400–411CrossRefPubMed Politis M, Loane C, Wu K, O’Sullivan SS, Woodhead Z, Kiferle L, Lawrence AD, Lees AJ, Piccini P (2013) Neural response to visual sexual cues in dopamine treatment-linked hypersexuality in Parkinson’s disease. Brain 136(Pt 2):400–411CrossRefPubMed
92.
go back to reference Ray NJ, Miyasaki JM, Zurowski M, Ko JH, Cho SS, Pellecchia G, Antonelli F, Houle S, Lang AE, Strafella AP (2012) Extrastriatal dopaminergic abnormalities of DA homeostasis in Parkinson’s patients with medication-induced pathological gambling: a [11C] FLB-457 and PET study. Neurobiol Dis 48(3):519–525PubMedCentralCrossRefPubMed Ray NJ, Miyasaki JM, Zurowski M, Ko JH, Cho SS, Pellecchia G, Antonelli F, Houle S, Lang AE, Strafella AP (2012) Extrastriatal dopaminergic abnormalities of DA homeostasis in Parkinson’s patients with medication-induced pathological gambling: a [11C] FLB-457 and PET study. Neurobiol Dis 48(3):519–525PubMedCentralCrossRefPubMed
93.
go back to reference Howland RH (2008) Understanding the placebo effect. Part 2: underlying psychological and neurobiological processes. J Psychosoc Nurs Ment Health Serv 46(6):15–18CrossRef Howland RH (2008) Understanding the placebo effect. Part 2: underlying psychological and neurobiological processes. J Psychosoc Nurs Ment Health Serv 46(6):15–18CrossRef
94.
go back to reference de la Fuente-Fernandez R (2009) The placebo-reward hypothesis: dopamine and the placebo effect. Parkinsonism Relat Disord 15(3):72–74CrossRef de la Fuente-Fernandez R (2009) The placebo-reward hypothesis: dopamine and the placebo effect. Parkinsonism Relat Disord 15(3):72–74CrossRef
95.
go back to reference Wolf S (1959) The pharmacology of placebos. J Am Med Assoc 159:1602–1606 Wolf S (1959) The pharmacology of placebos. J Am Med Assoc 159:1602–1606
96.
go back to reference Shetty N, Friedman JH, Kieburtz K, Marshall FJ, Oakes D (1999) The placebo response in Parkinson’s disease. Parkinson Study Group. Clin Neuropharmacol 22(4):207–212PubMed Shetty N, Friedman JH, Kieburtz K, Marshall FJ, Oakes D (1999) The placebo response in Parkinson’s disease. Parkinson Study Group. Clin Neuropharmacol 22(4):207–212PubMed
97.
go back to reference de la Fuente-Fernandez R, Ruth TJ, Sossi V, Schulzer M, Calne DB, Stoessl AJ (2001) Expectation and dopamine release: mechanisms of the placebo effect in Parkinson’s disease. Science 293:1164–1166CrossRefPubMed de la Fuente-Fernandez R, Ruth TJ, Sossi V, Schulzer M, Calne DB, Stoessl AJ (2001) Expectation and dopamine release: mechanisms of the placebo effect in Parkinson’s disease. Science 293:1164–1166CrossRefPubMed
98.
go back to reference de la Fuente-Fernández R, Phillips AG, Zamburlini M, Sossi V, Calne DB, Ruth TJ, Stoessl AJ (2002) Dopamine release in human ventral striatum and expectation of reward. Behav Brain Res 136(2):359–363CrossRefPubMed de la Fuente-Fernández R, Phillips AG, Zamburlini M, Sossi V, Calne DB, Ruth TJ, Stoessl AJ (2002) Dopamine release in human ventral striatum and expectation of reward. Behav Brain Res 136(2):359–363CrossRefPubMed
99.
go back to reference Lidstone SC, Schulzer M, Dinelle K, Mak E, Sossi V, Ruth TJ, de la Fuente-Fernández R, Phillips AG, Stoessl AJ (2010) Effects of expectation on placebo-induced dopamine release in Parkinson disease. Arch Gen Psychiatry 67(8):857–865CrossRefPubMed Lidstone SC, Schulzer M, Dinelle K, Mak E, Sossi V, Ruth TJ, de la Fuente-Fernández R, Phillips AG, Stoessl AJ (2010) Effects of expectation on placebo-induced dopamine release in Parkinson disease. Arch Gen Psychiatry 67(8):857–865CrossRefPubMed
100.
go back to reference Fisher BE, Li Q, Nacca A, Salem GJ, Song J, Yip J, Hui JS, Jakowec MW, Petzinger GM (2013) Treadmill exercise elevates striatal dopamine D2 receptor binding potential in patients with early Parkinson’s disease. Neuroreport 24(10):509–514CrossRefPubMed Fisher BE, Li Q, Nacca A, Salem GJ, Song J, Yip J, Hui JS, Jakowec MW, Petzinger GM (2013) Treadmill exercise elevates striatal dopamine D2 receptor binding potential in patients with early Parkinson’s disease. Neuroreport 24(10):509–514CrossRefPubMed
Metadata
Title
Dopamine receptor mapping with PET imaging in Parkinson’s disease
Authors
Flavia Niccolini
Paul Su
Marios Politis
Publication date
01-12-2014
Publisher
Springer Berlin Heidelberg
Published in
Journal of Neurology / Issue 12/2014
Print ISSN: 0340-5354
Electronic ISSN: 1432-1459
DOI
https://doi.org/10.1007/s00415-014-7302-2

Other articles of this Issue 12/2014

Journal of Neurology 12/2014 Go to the issue