Skip to main content
Top
Published in: Translational Neurodegeneration 1/2017

Open Access 01-12-2017 | Review

Imaging biomarkers in Parkinson’s disease and Parkinsonian syndromes: current and emerging concepts

Authors: Usman Saeed, Jordana Compagnone, Richard I. Aviv, Antonio P. Strafella, Sandra E. Black, Anthony E. Lang, Mario Masellis

Published in: Translational Neurodegeneration | Issue 1/2017

Login to get access

Abstract

Two centuries ago in 1817, James Parkinson provided the first medical description of Parkinson’s disease, later refined by Jean-Martin Charcot in the mid-to-late 19th century to include the atypical parkinsonian variants (also termed, Parkinson-plus syndromes). Today, Parkinson’s disease represents the second most common neurodegenerative disorder with an estimated global prevalence of over 10 million. Conversely, atypical parkinsonian syndromes encompass a group of relatively heterogeneous disorders that may share some clinical features with Parkinson’s disease, but are uncommon distinct clinicopathological diseases. Decades of scientific advancements have vastly improved our understanding of these disorders, including improvements in in vivo imaging for biomarker identification. Multimodal imaging for the visualization of structural and functional brain changes is especially important, as it allows a ‘window’ into the underlying pathophysiological abnormalities. In this article, we first present an overview of the cardinal clinical and neuropathological features of, 1) synucleinopathies: Parkinson’s disease and other Lewy body spectrum disorders, as well as multiple system atrophy, and 2) tauopathies: progressive supranuclear palsy, and corticobasal degeneration. A comprehensive presentation of well-established and emerging imaging biomarkers for each disorder are then discussed. Biomarkers for the following imaging modalities are reviewed: 1) structural magnetic resonance imaging (MRI) using T1, T2, and susceptibility-weighted sequences for volumetric and voxel-based morphometric analyses, as well as MRI derived visual signatures, 2) diffusion tensor MRI for the assessment of white matter tract injury and microstructural integrity, 3) proton magnetic resonance spectroscopy for quantifying proton-containing brain metabolites, 4) single photon emission computed tomography for the evaluation of nigrostriatal integrity (as assessed by presynaptic dopamine transporters and postsynaptic dopamine D2 receptors), and cerebral perfusion, 5) positron emission tomography for gauging nigrostriatal functions, glucose metabolism, amyloid and tau molecular imaging, as well as neuroinflammation, 6) myocardial scintigraphy for dysautonomia, and 7) transcranial sonography for measuring substantia nigra and lentiform nucleus echogenicity. Imaging biomarkers, using the ‘multimodal approach’, may aid in making early, accurate and objective diagnostic decisions, highlight neuroanatomical and pathophysiological mechanisms, as well as assist in evaluating disease progression and therapeutic responses to drugs in clinical trials.
Literature
2.
4.
go back to reference McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB consortium. Neurology. 2005;65:1863–72.PubMedCrossRef McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB consortium. Neurology. 2005;65:1863–72.PubMedCrossRef
7.
go back to reference Dickson DW, Rademakers R, Hutton ML. Progressive Supranuclear palsy: pathology and genetics. Brain Pathol. 2007;17:74–82.PubMedCrossRef Dickson DW, Rademakers R, Hutton ML. Progressive Supranuclear palsy: pathology and genetics. Brain Pathol. 2007;17:74–82.PubMedCrossRef
8.
go back to reference Williams DR, Lees AJ. Progressive Supranuclear palsy: clinicopathological concepts and diagnostic challenges. Lancet Neurol. 2009;8:270–79.PubMedCrossRef Williams DR, Lees AJ. Progressive Supranuclear palsy: clinicopathological concepts and diagnostic challenges. Lancet Neurol. 2009;8:270–79.PubMedCrossRef
9.
go back to reference Armstrong MJ, Litvan I, Lang AE, Bak TH, Bhatia KP, Borroni B, et al. Criteria for the diagnosis of corticobasal degeneration. Neurology. 2013;80:496–503.PubMedPubMedCentralCrossRef Armstrong MJ, Litvan I, Lang AE, Bak TH, Bhatia KP, Borroni B, et al. Criteria for the diagnosis of corticobasal degeneration. Neurology. 2013;80:496–503.PubMedPubMedCentralCrossRef
10.
go back to reference Litvan I, Agid Y, Goetz C, Jankovic J, Wenning GK, Brandel JP, et al. Accuracy of the clinical diagnosis of corticobasal degeneration: a clinicopathologic study. Neurology. 1997;48:119–25.PubMedCrossRef Litvan I, Agid Y, Goetz C, Jankovic J, Wenning GK, Brandel JP, et al. Accuracy of the clinical diagnosis of corticobasal degeneration: a clinicopathologic study. Neurology. 1997;48:119–25.PubMedCrossRef
11.
go back to reference Whitwell JL, Jack CR, Boeve BF, Parisi JE, Ahlskog JE, Drubach DA, et al. Imaging correlates of pathology in corticobasal syndrome. Neurology. 2010;75:1879–87.PubMedPubMedCentralCrossRef Whitwell JL, Jack CR, Boeve BF, Parisi JE, Ahlskog JE, Drubach DA, et al. Imaging correlates of pathology in corticobasal syndrome. Neurology. 2010;75:1879–87.PubMedPubMedCentralCrossRef
12.
go back to reference Burton EJ, McKeith IG, Burn DJ, Williams ED, O’Brien JT. Cerebral atrophy in Parkinson’s disease with and without dementia: a comparison with Alzheimer’s disease, dementia with Lewy bodies and controls. Brain. 2004;127:791–800.PubMedCrossRef Burton EJ, McKeith IG, Burn DJ, Williams ED, O’Brien JT. Cerebral atrophy in Parkinson’s disease with and without dementia: a comparison with Alzheimer’s disease, dementia with Lewy bodies and controls. Brain. 2004;127:791–800.PubMedCrossRef
13.
go back to reference Summerfield C, Junqué C, Tolosa E, Salgado-Pineda P, Gómez-Ansón B, Martí MJ, et al. Structural brain changes in Parkinson disease with dementia: a voxel-based morphometry study. Arch Neurol. 2005;62:281–85.PubMedCrossRef Summerfield C, Junqué C, Tolosa E, Salgado-Pineda P, Gómez-Ansón B, Martí MJ, et al. Structural brain changes in Parkinson disease with dementia: a voxel-based morphometry study. Arch Neurol. 2005;62:281–85.PubMedCrossRef
14.
go back to reference Pitcher TL, Melzer TR, MacAskill MR, Graham CF, Livingston L, Keenan RJ, et al. Reduced Striatal volumes in Parkinson’s disease: a magnetic resonance imaging study. Transl Neurodegener. 2012;1:1–8.CrossRef Pitcher TL, Melzer TR, MacAskill MR, Graham CF, Livingston L, Keenan RJ, et al. Reduced Striatal volumes in Parkinson’s disease: a magnetic resonance imaging study. Transl Neurodegener. 2012;1:1–8.CrossRef
15.
go back to reference Tinaz S, Courtney MG, Stern CE. Focal cortical and subcortical atrophy in early Parkinson’s disease. Mov Disord. 2011;26:436–41.PubMedCrossRef Tinaz S, Courtney MG, Stern CE. Focal cortical and subcortical atrophy in early Parkinson’s disease. Mov Disord. 2011;26:436–41.PubMedCrossRef
16.
go back to reference Schulz JB, Skalej M, Wedekind D, Luft AR, Abele M, Voigt K, et al. Magnetic resonance imaging-based volumetry differentiates idiopathic Parkinson’s syndrome from multiple system atrophy and progressive Supranuclear palsy. Ann Neurol. 1999;45:65–74.PubMedCrossRef Schulz JB, Skalej M, Wedekind D, Luft AR, Abele M, Voigt K, et al. Magnetic resonance imaging-based volumetry differentiates idiopathic Parkinson’s syndrome from multiple system atrophy and progressive Supranuclear palsy. Ann Neurol. 1999;45:65–74.PubMedCrossRef
17.
go back to reference Chen S, Tan HY, Wu ZH, Sun CP, He JX, Li XC, et al. Imaging of olfactory bulb and gray matter volumes in brain areas associated with olfactory function in patients with Parkinson’s disease and multiple system atrophy. Eur J Radiol. 2014;83:564–70.PubMedCrossRef Chen S, Tan HY, Wu ZH, Sun CP, He JX, Li XC, et al. Imaging of olfactory bulb and gray matter volumes in brain areas associated with olfactory function in patients with Parkinson’s disease and multiple system atrophy. Eur J Radiol. 2014;83:564–70.PubMedCrossRef
18.
go back to reference Oikawa H, Sasaki M, Tamakawa Y, Ehara S, Tohyama K. The substantia nigra in Parkinson disease: proton density-weighted spin-echo and fast short inversion time inversion-recovery MR findings. Am J Neuroradiol. 2002;23:1747–56.PubMed Oikawa H, Sasaki M, Tamakawa Y, Ehara S, Tohyama K. The substantia nigra in Parkinson disease: proton density-weighted spin-echo and fast short inversion time inversion-recovery MR findings. Am J Neuroradiol. 2002;23:1747–56.PubMed
19.
go back to reference Péran P, Cherubini A, Assogna F, Piras F, Quattrocchi C, Peppe A, et al. Magnetic resonance imaging markers of Parkinson’s disease nigrostriatal signature. Brain. 2010;133:3423–33.PubMedCrossRef Péran P, Cherubini A, Assogna F, Piras F, Quattrocchi C, Peppe A, et al. Magnetic resonance imaging markers of Parkinson’s disease nigrostriatal signature. Brain. 2010;133:3423–33.PubMedCrossRef
20.
go back to reference Minati L, Grisoli M, Carella F, De Simone T, Bruzzone MG, Savoiardo M. Imaging degeneration of the substantia nigra in Parkinson disease with inversion-recovery MR imaging. Am J Neuroradiol. 2007;28:309–13.PubMed Minati L, Grisoli M, Carella F, De Simone T, Bruzzone MG, Savoiardo M. Imaging degeneration of the substantia nigra in Parkinson disease with inversion-recovery MR imaging. Am J Neuroradiol. 2007;28:309–13.PubMed
21.
go back to reference Kwon DH, Kim JM, Oh SH, Jeong HJ, Park SY, Oh ES, et al. Seven-tesla magnetic resonance images of the substantia nigra in Parkinson disease. Ann Neurol. 2012;71:267–77.PubMedCrossRef Kwon DH, Kim JM, Oh SH, Jeong HJ, Park SY, Oh ES, et al. Seven-tesla magnetic resonance images of the substantia nigra in Parkinson disease. Ann Neurol. 2012;71:267–77.PubMedCrossRef
22.
go back to reference Cho ZH, Oh SH, Kim JM, Park SY, Kwon DH, Jeong HJ, et al. Direct visualization of Parkinson’s disease by in vivo human brain imaging using 7.0 T magnetic resonance imaging. Mov Disord. 2011;26:713–8.PubMedCrossRef Cho ZH, Oh SH, Kim JM, Park SY, Kwon DH, Jeong HJ, et al. Direct visualization of Parkinson’s disease by in vivo human brain imaging using 7.0 T magnetic resonance imaging. Mov Disord. 2011;26:713–8.PubMedCrossRef
23.
go back to reference Cosottini M, Frosini D, Pesaresi I, Costagli M, Biagi L, Ceravolo R, et al. MR imaging of the substantia nigra at 7 T enables diagnosis of Parkinson disease. Radiology. 2014;271:831–8.PubMedCrossRef Cosottini M, Frosini D, Pesaresi I, Costagli M, Biagi L, Ceravolo R, et al. MR imaging of the substantia nigra at 7 T enables diagnosis of Parkinson disease. Radiology. 2014;271:831–8.PubMedCrossRef
24.
go back to reference Ulla M, Bonny JM, Ouchchane L, Rieu I, Claise B, Durif F. Is R2* a New MRI biomarker for the progression of Parkinson’s disease? a longitudinal follow-Up. PLoS ONE. 2013;8:e57904.PubMedPubMedCentralCrossRef Ulla M, Bonny JM, Ouchchane L, Rieu I, Claise B, Durif F. Is R2* a New MRI biomarker for the progression of Parkinson’s disease? a longitudinal follow-Up. PLoS ONE. 2013;8:e57904.PubMedPubMedCentralCrossRef
25.
go back to reference Wieler M, Gee M, Camicioli R, Martin WRW. Freezing of gait in early Parkinson‘s disease: nigral iron content estimated from magnetic resonance imaging. J Neurol Sci. 2016;361:87–91.PubMedCrossRef Wieler M, Gee M, Camicioli R, Martin WRW. Freezing of gait in early Parkinson‘s disease: nigral iron content estimated from magnetic resonance imaging. J Neurol Sci. 2016;361:87–91.PubMedCrossRef
26.
go back to reference Damier P, Hirsch EC, Agid Y, Graybiel AM. The substantia nigra of the human brain: I. Nigrosomes and the nigral matrix, a compartmental organization based on calbindin D(28 K) immunohistochemistry. Brain. 1999;122:1421–36.PubMedCrossRef Damier P, Hirsch EC, Agid Y, Graybiel AM. The substantia nigra of the human brain: I. Nigrosomes and the nigral matrix, a compartmental organization based on calbindin D(28 K) immunohistochemistry. Brain. 1999;122:1421–36.PubMedCrossRef
27.
go back to reference Damier P, Hirsch EC, Agid Y, Graybiel AM. The substantia nigra of the human brain: II. Patterns of loss of dopamine-containing neurons in Parkinson’s disease. Brain. 1999;122:1437–48.PubMedCrossRef Damier P, Hirsch EC, Agid Y, Graybiel AM. The substantia nigra of the human brain: II. Patterns of loss of dopamine-containing neurons in Parkinson’s disease. Brain. 1999;122:1437–48.PubMedCrossRef
28.
go back to reference Blazejewska AI, Schwarz ST, Pitiot A, Stephenson MC, Lowe J, Bajaj N, et al. Visualization of nigrosome 1 and its loss in PD: pathoanatomical correlation and in vivo 7 T MRI. Neurology. 2013;81:534–40.PubMedPubMedCentralCrossRef Blazejewska AI, Schwarz ST, Pitiot A, Stephenson MC, Lowe J, Bajaj N, et al. Visualization of nigrosome 1 and its loss in PD: pathoanatomical correlation and in vivo 7 T MRI. Neurology. 2013;81:534–40.PubMedPubMedCentralCrossRef
29.
go back to reference Lehéricy S, Bardinet E, Poupon C, Vidailhet M, François C. 7 tesla magnetic resonance imaging: a closer look at substantia nigra anatomy in Parkinson’s disease. Mov Disord. 2014;29:1574–81.PubMedCrossRef Lehéricy S, Bardinet E, Poupon C, Vidailhet M, François C. 7 tesla magnetic resonance imaging: a closer look at substantia nigra anatomy in Parkinson’s disease. Mov Disord. 2014;29:1574–81.PubMedCrossRef
30.
go back to reference Castellanos G, Fernández-Seara MA, Lorenzo-Betancor O, Ortega-Cubero S, Puigvert M, Uranga J, et al. Automated neuromelanin imaging as a diagnostic biomarker for Parkinson’s disease. Mov Disord. 2015;30:945–52.PubMedCrossRef Castellanos G, Fernández-Seara MA, Lorenzo-Betancor O, Ortega-Cubero S, Puigvert M, Uranga J, et al. Automated neuromelanin imaging as a diagnostic biomarker for Parkinson’s disease. Mov Disord. 2015;30:945–52.PubMedCrossRef
31.
go back to reference Reimão S, Pita Lobo P, Neutel D, Guedes LC, Coelho M, Rosa MM, et al. Substantia nigra neuromelanin-MR imaging differentiates essential tremor from Parkinson’s disease. Mov Disord. 2015;30:953–9.PubMedCrossRef Reimão S, Pita Lobo P, Neutel D, Guedes LC, Coelho M, Rosa MM, et al. Substantia nigra neuromelanin-MR imaging differentiates essential tremor from Parkinson’s disease. Mov Disord. 2015;30:953–9.PubMedCrossRef
32.
go back to reference Schwarz ST, Afzal M, Morgan PS, Bajaj N, Gowland PA, Auer DP. The “swallow tail” appearance of the healthy nigrosome - a new accurate test of Parkinson’s disease: a case–control and retrospective cross-sectional MRI study at 3 T. PLoS ONE. 2014;9:e93814.PubMedPubMedCentralCrossRef Schwarz ST, Afzal M, Morgan PS, Bajaj N, Gowland PA, Auer DP. The “swallow tail” appearance of the healthy nigrosome - a new accurate test of Parkinson’s disease: a case–control and retrospective cross-sectional MRI study at 3 T. PLoS ONE. 2014;9:e93814.PubMedPubMedCentralCrossRef
33.
go back to reference Beyer MK, Larsen JP, Aarsland D. Gray matter atrophy in Parkinson disease with dementia and dementia with Lewy bodies. Neurology. 2007;69:747–54.PubMedCrossRef Beyer MK, Larsen JP, Aarsland D. Gray matter atrophy in Parkinson disease with dementia and dementia with Lewy bodies. Neurology. 2007;69:747–54.PubMedCrossRef
34.
go back to reference Ballard C, Ziabreva I, Perry R, Larsen JP, O’Brien J, McKeith I, et al. Differences in neuropathologic characteristics across the Lewy body dementia spectrum. Neurology. 2006;67:1931–4.PubMedCrossRef Ballard C, Ziabreva I, Perry R, Larsen JP, O’Brien J, McKeith I, et al. Differences in neuropathologic characteristics across the Lewy body dementia spectrum. Neurology. 2006;67:1931–4.PubMedCrossRef
35.
go back to reference Goldman JG, Stebbins GT, Bernard B, Stoub TR, Goetz CG, deToledo-Morrell L. Entorhinal cortex atrophy differentiates Parkinson’s disease patients with and without dementia. Mov Disord. 2012;27:727–34.PubMedPubMedCentralCrossRef Goldman JG, Stebbins GT, Bernard B, Stoub TR, Goetz CG, deToledo-Morrell L. Entorhinal cortex atrophy differentiates Parkinson’s disease patients with and without dementia. Mov Disord. 2012;27:727–34.PubMedPubMedCentralCrossRef
36.
go back to reference Camicioli R, Moore MM, Kinney A, Corbridge E, Glassberg K, Kaye JA. Parkinson’s disease is associated with Hippocampal atrophy. Mov Disord. 2003;18:784–90.PubMedCrossRef Camicioli R, Moore MM, Kinney A, Corbridge E, Glassberg K, Kaye JA. Parkinson’s disease is associated with Hippocampal atrophy. Mov Disord. 2003;18:784–90.PubMedCrossRef
37.
go back to reference Cousins DA, Burton EJ, Burn D, Gholkar A, McKeith IG, O’Brien JT. Atrophy of the putamen in dementia with Lewy bodies but not Alzheimer’s disease: an MRI study. Neurology. 2003;61:1191–5.PubMedCrossRef Cousins DA, Burton EJ, Burn D, Gholkar A, McKeith IG, O’Brien JT. Atrophy of the putamen in dementia with Lewy bodies but not Alzheimer’s disease: an MRI study. Neurology. 2003;61:1191–5.PubMedCrossRef
38.
go back to reference Beyer MK, Aarsland D, Greve OJ, Larsen JP. Visual rating of white matter hyperintensities in Parkinson’s disease. Mov Disord. 2006;21:223–9.PubMedCrossRef Beyer MK, Aarsland D, Greve OJ, Larsen JP. Visual rating of white matter hyperintensities in Parkinson’s disease. Mov Disord. 2006;21:223–9.PubMedCrossRef
39.
go back to reference Mak E, Bergsland N, Dwyer MG, Zivadinov R, Kandiah N. Subcortical atrophy is associated with cognitive impairment in mild Parkinson disease: a combined investigation of volumetric changes, cortical thickness, and vertex-based shape analysis. Am J Neuroradiol. 2014;35:2257–64.PubMedCrossRef Mak E, Bergsland N, Dwyer MG, Zivadinov R, Kandiah N. Subcortical atrophy is associated with cognitive impairment in mild Parkinson disease: a combined investigation of volumetric changes, cortical thickness, and vertex-based shape analysis. Am J Neuroradiol. 2014;35:2257–64.PubMedCrossRef
40.
go back to reference Hanganu A, Bedetti C, Degroot C, Mejia-Constain B, Lafontaine AL, Soland V, et al. Mild cognitive impairment is linked with faster rate of cortical thinning in patients with Parkinson’s disease longitudinally. Brain. 2014;137:1120–9.PubMedCrossRef Hanganu A, Bedetti C, Degroot C, Mejia-Constain B, Lafontaine AL, Soland V, et al. Mild cognitive impairment is linked with faster rate of cortical thinning in patients with Parkinson’s disease longitudinally. Brain. 2014;137:1120–9.PubMedCrossRef
41.
go back to reference Monchi O, Hanganu A, Bellec P. Markers of cognitive decline in PD: the case for heterogeneity. Park Relat Disord. 2016;24:8–14.CrossRef Monchi O, Hanganu A, Bellec P. Markers of cognitive decline in PD: the case for heterogeneity. Park Relat Disord. 2016;24:8–14.CrossRef
42.
go back to reference Weintraub D, Dietz N, Duda JE, Wolk DA, Doshi J, Xie SX, et al. Alzheimer’s disease pattern of brain atrophy predicts cognitive decline in Parkinson’s disease. Brain. 2012;135:170–80.PubMedCrossRef Weintraub D, Dietz N, Duda JE, Wolk DA, Doshi J, Xie SX, et al. Alzheimer’s disease pattern of brain atrophy predicts cognitive decline in Parkinson’s disease. Brain. 2012;135:170–80.PubMedCrossRef
43.
go back to reference Massey LA, Micallef C, Paviour DC, O’Sullivan SS, Ling H, Williams DR, et al. Conventional magnetic resonance imaging in confirmed progressive Supranuclear palsy and multiple system atrophy. Mov Disord. 2012;27:1754–62.PubMedCrossRef Massey LA, Micallef C, Paviour DC, O’Sullivan SS, Ling H, Williams DR, et al. Conventional magnetic resonance imaging in confirmed progressive Supranuclear palsy and multiple system atrophy. Mov Disord. 2012;27:1754–62.PubMedCrossRef
44.
go back to reference Feng J, Huang B, Yang W, Zhang Y, Wang L, Wang L, et al. The putaminal abnormalities on 3.0 T magnetic resonance imaging: can they separate parkinsonism-predominant multiple system atrophy from Parkinson’s disease? Acta Radiol. 2014;56:322–8.PubMedCrossRef Feng J, Huang B, Yang W, Zhang Y, Wang L, Wang L, et al. The putaminal abnormalities on 3.0 T magnetic resonance imaging: can they separate parkinsonism-predominant multiple system atrophy from Parkinson’s disease? Acta Radiol. 2014;56:322–8.PubMedCrossRef
45.
go back to reference Sako W, Murakami N, Izumi Y, Kaji R. The difference in putamen volume between {MSA} and PD: evidence from a meta-analysis. Parkinsonism Relat Disord. 2014;20:873–7.PubMedCrossRef Sako W, Murakami N, Izumi Y, Kaji R. The difference in putamen volume between {MSA} and PD: evidence from a meta-analysis. Parkinsonism Relat Disord. 2014;20:873–7.PubMedCrossRef
46.
go back to reference Deguchi K, Ikeda K, Kume K, Takata T, Kokudo Y, Kamada M, et al. Significance of the hot-cross bun sign on T2*-weighted MRI for the diagnosis of multiple system atrophy. J Neurol. 2015;262(6):1433–9.PubMedCrossRef Deguchi K, Ikeda K, Kume K, Takata T, Kokudo Y, Kamada M, et al. Significance of the hot-cross bun sign on T2*-weighted MRI for the diagnosis of multiple system atrophy. J Neurol. 2015;262(6):1433–9.PubMedCrossRef
47.
go back to reference Wadia PM, Howard P, Ribeirro MQ, Robblee J, Asante A, Mikulis DJ, et al. The value of GRE, ADC and routine MRI in distinguishing Parkinsonian disorders. Can J Neurol Sci. 2013;40:389–402.PubMedCrossRef Wadia PM, Howard P, Ribeirro MQ, Robblee J, Asante A, Mikulis DJ, et al. The value of GRE, ADC and routine MRI in distinguishing Parkinsonian disorders. Can J Neurol Sci. 2013;40:389–402.PubMedCrossRef
48.
go back to reference Tir M, Delmaire C, le Thuc V, Duhamel A, Destée A, Pruvo JP, et al. Motor-related circuit dysfunction in MSA-P: usefulness of combined whole-brain imaging analysis. Mov Disord. 2009;24:863–70.PubMedCrossRef Tir M, Delmaire C, le Thuc V, Duhamel A, Destée A, Pruvo JP, et al. Motor-related circuit dysfunction in MSA-P: usefulness of combined whole-brain imaging analysis. Mov Disord. 2009;24:863–70.PubMedCrossRef
49.
go back to reference Brenneis C, Seppi K, Schocke MF, Müller J, Luginger E, Bösch S, et al. Voxel-based morphometry detects cortical atrophy in the Parkinson variant of multiple system atrophy. Mov Disord. 2003;18:1132–8.PubMedCrossRef Brenneis C, Seppi K, Schocke MF, Müller J, Luginger E, Bösch S, et al. Voxel-based morphometry detects cortical atrophy in the Parkinson variant of multiple system atrophy. Mov Disord. 2003;18:1132–8.PubMedCrossRef
50.
go back to reference Kim HJ, Jeon BS, Kim YE, Kim JY, Kim YK, Sohn CH, et al. Clinical and imaging characteristics of dementia in multiple system atrophy. Parkinsonism Relat Disord. 2013;19:617–21.PubMedCrossRef Kim HJ, Jeon BS, Kim YE, Kim JY, Kim YK, Sohn CH, et al. Clinical and imaging characteristics of dementia in multiple system atrophy. Parkinsonism Relat Disord. 2013;19:617–21.PubMedCrossRef
51.
go back to reference Reginold W, Lang AE, Marras C, Heyn C, Alharbi M, Mikulis DJ. Longitudinal quantitative MRI in multiple system atrophy and progressive Supranuclear palsy. Parkinsonism Relat Disord. 2014;20:222–5.PubMedCrossRef Reginold W, Lang AE, Marras C, Heyn C, Alharbi M, Mikulis DJ. Longitudinal quantitative MRI in multiple system atrophy and progressive Supranuclear palsy. Parkinsonism Relat Disord. 2014;20:222–5.PubMedCrossRef
52.
go back to reference Focke NK, Helms G, Pantel PM, Scheewe S, Knauth M, Bachmann CG, et al. Differentiation of typical and atypical Parkinson syndromes by quantitative MR imaging. Am J Neuroradiol. 2011;32:2087–92.PubMedCrossRef Focke NK, Helms G, Pantel PM, Scheewe S, Knauth M, Bachmann CG, et al. Differentiation of typical and atypical Parkinson syndromes by quantitative MR imaging. Am J Neuroradiol. 2011;32:2087–92.PubMedCrossRef
53.
go back to reference Wang Y, Butros SR, Shuai X, Dai Y, Chen C, Liu M, et al. Different iron-deposition patterns of multiple system atrophy with predominant parkinsonism and idiopathetic Parkinson diseases demonstrated by phase-corrected susceptibility-weighted imaging. Am J Neuroradiol. 2012;33:266–73.PubMedCrossRef Wang Y, Butros SR, Shuai X, Dai Y, Chen C, Liu M, et al. Different iron-deposition patterns of multiple system atrophy with predominant parkinsonism and idiopathetic Parkinson diseases demonstrated by phase-corrected susceptibility-weighted imaging. Am J Neuroradiol. 2012;33:266–73.PubMedCrossRef
54.
go back to reference Boxer AL, Geschwind MD, Belfor N, Gorno-Tempini ML, Schauer GF, Miller BL, et al. Patterns of brain atrophy that differentiate corticobasal degeneration syndrome from progressive supranuclear palsy. Arch Neurol. 2006;63:81–6.PubMedCrossRef Boxer AL, Geschwind MD, Belfor N, Gorno-Tempini ML, Schauer GF, Miller BL, et al. Patterns of brain atrophy that differentiate corticobasal degeneration syndrome from progressive supranuclear palsy. Arch Neurol. 2006;63:81–6.PubMedCrossRef
55.
go back to reference Kurata T, Kametaka S, Ohta Y, Morimoto N, Deguchi S, Deguchi K, et al. PSP as distinguished from CBD, MSA-P and PD by clinical and imaging differences at an early stage. Intern Med. 2011;50:2775–81.PubMedCrossRef Kurata T, Kametaka S, Ohta Y, Morimoto N, Deguchi S, Deguchi K, et al. PSP as distinguished from CBD, MSA-P and PD by clinical and imaging differences at an early stage. Intern Med. 2011;50:2775–81.PubMedCrossRef
56.
go back to reference Gröschel K, Hauser TK, Luft A, Patronas N, Dichgans J, Litvan I, et al. Magnetic resonance imaging-based volumetry differentiates progressive supranuclear palsy from corticobasal degeneration. Neuroimage. 2004;21:714–24.PubMedCrossRef Gröschel K, Hauser TK, Luft A, Patronas N, Dichgans J, Litvan I, et al. Magnetic resonance imaging-based volumetry differentiates progressive supranuclear palsy from corticobasal degeneration. Neuroimage. 2004;21:714–24.PubMedCrossRef
57.
go back to reference Brenneis C, Seppi K, Schocke M, Benke T, Wenning GK, Poewe W. Voxel based morphometry reveals a distinct pattern of frontal atrophy in progressive supranuclear palsy. J Neurol Neurosurg Psychiatry. 2004;75:246–9.PubMedPubMedCentral Brenneis C, Seppi K, Schocke M, Benke T, Wenning GK, Poewe W. Voxel based morphometry reveals a distinct pattern of frontal atrophy in progressive supranuclear palsy. J Neurol Neurosurg Psychiatry. 2004;75:246–9.PubMedPubMedCentral
58.
go back to reference Padovani A, Borroni B, Brambati SM, Agosti C, Broli M, Alonso R, et al. Diffusion tensor imaging and voxel based morphometry study in early progressive supranuclear palsy. J Neurol Neurosurg Psychiatry. 2006;77:457–63.PubMedCrossRef Padovani A, Borroni B, Brambati SM, Agosti C, Broli M, Alonso R, et al. Diffusion tensor imaging and voxel based morphometry study in early progressive supranuclear palsy. J Neurol Neurosurg Psychiatry. 2006;77:457–63.PubMedCrossRef
59.
go back to reference Price S, Paviour D, Scahill R, Stevens J, Rossor M, Lees A, et al. Voxel-based morphometry detects patterns of atrophy that help differentiate progressive supranuclear palsy and Parkinson’s disease. Neuroimage. 2004;23:663–9.PubMedCrossRef Price S, Paviour D, Scahill R, Stevens J, Rossor M, Lees A, et al. Voxel-based morphometry detects patterns of atrophy that help differentiate progressive supranuclear palsy and Parkinson’s disease. Neuroimage. 2004;23:663–9.PubMedCrossRef
60.
go back to reference Paviour DC, Price SL, Jahanshahi M, Lees AJ, Fox NC. Longitudinal MRI in progressive supranuclear palsy and multiple system atrophy: rates and regions of atrophy. Brain. 2006;129:1040–9.PubMedCrossRef Paviour DC, Price SL, Jahanshahi M, Lees AJ, Fox NC. Longitudinal MRI in progressive supranuclear palsy and multiple system atrophy: rates and regions of atrophy. Brain. 2006;129:1040–9.PubMedCrossRef
61.
go back to reference Josephs KA, Whitwell JL, Dickson DW, Boeve BF, Knopman DS, Petersen RC, et al. Voxel-based morphometry in autopsy proven PSP and CBD. Neurobiol Aging. 2008;29:280–9.PubMedCrossRef Josephs KA, Whitwell JL, Dickson DW, Boeve BF, Knopman DS, Petersen RC, et al. Voxel-based morphometry in autopsy proven PSP and CBD. Neurobiol Aging. 2008;29:280–9.PubMedCrossRef
62.
go back to reference Quattrone A, Nicoletti G, Messina D, Fera F, Condino F, Pugliese P, et al. MR imaging index for differentiation of progressive supranuclear palsy from Parkinson disease and the Parkinson variant of multiple system atrophy. Radiology. 2008;246:214–21.PubMedCrossRef Quattrone A, Nicoletti G, Messina D, Fera F, Condino F, Pugliese P, et al. MR imaging index for differentiation of progressive supranuclear palsy from Parkinson disease and the Parkinson variant of multiple system atrophy. Radiology. 2008;246:214–21.PubMedCrossRef
63.
go back to reference Kaasinen V, Kangassalo N, Gardberg M, Isotalo J, Karhu J, Parkkola R, et al. Midbrain-to-pons ratio in autopsy-confirmed progressive supranuclear palsy: replication in an independent cohort. Neurol Sci. 2015;36:1251–3.PubMedCrossRef Kaasinen V, Kangassalo N, Gardberg M, Isotalo J, Karhu J, Parkkola R, et al. Midbrain-to-pons ratio in autopsy-confirmed progressive supranuclear palsy: replication in an independent cohort. Neurol Sci. 2015;36:1251–3.PubMedCrossRef
64.
go back to reference Whitwell JL, Xu J, Mandrekar JN, Gunter JL, Jack CR, Josephs KA. Rates of brain atrophy and clinical decline over 6 and 12-month intervals in PSP: Determining sample size for treatment trials. Park Relat Disord. 2012;18:252–6.CrossRef Whitwell JL, Xu J, Mandrekar JN, Gunter JL, Jack CR, Josephs KA. Rates of brain atrophy and clinical decline over 6 and 12-month intervals in PSP: Determining sample size for treatment trials. Park Relat Disord. 2012;18:252–6.CrossRef
65.
go back to reference Borroni B, Garibotto V, Agosti C, Brambati SM, Bellelli G, Gasparotti R, et al. White matter changes in corticobasal degeneration syndrome and correlation with limb apraxia. Arch Neurol. 2008;65:796–801.PubMedCrossRef Borroni B, Garibotto V, Agosti C, Brambati SM, Bellelli G, Gasparotti R, et al. White matter changes in corticobasal degeneration syndrome and correlation with limb apraxia. Arch Neurol. 2008;65:796–801.PubMedCrossRef
66.
go back to reference Schofield EC, Caine D, Kril JJ, Cordato NJ, Halliday GM. Staging disease severity in movement disorder tauopathies: brain atrophy separates progressive Supranuclear palsy from corticobasal degeneration. Mov Disord. 2005;20:34–9.PubMedCrossRef Schofield EC, Caine D, Kril JJ, Cordato NJ, Halliday GM. Staging disease severity in movement disorder tauopathies: brain atrophy separates progressive Supranuclear palsy from corticobasal degeneration. Mov Disord. 2005;20:34–9.PubMedCrossRef
67.
go back to reference Josephs KA, Tang-Wai DF, Edland SD, Knopman DS, Dickson DW, Parisi JE, et al. Correlation between antemortem magnetic resonance imaging findings and pathologically confirmed corticobasal degeneration. Arch Neurol. 2004;61:1881–4.PubMed Josephs KA, Tang-Wai DF, Edland SD, Knopman DS, Dickson DW, Parisi JE, et al. Correlation between antemortem magnetic resonance imaging findings and pathologically confirmed corticobasal degeneration. Arch Neurol. 2004;61:1881–4.PubMed
68.
go back to reference Lee SE, Rabinovici GD, Mayo MC, Wilson SM, Seeley WW, Dearmond SJ, et al. Clinicopathological correlations in corticobasal degeneration. Ann Neurol. 2011;70:327–40.PubMedPubMedCentralCrossRef Lee SE, Rabinovici GD, Mayo MC, Wilson SM, Seeley WW, Dearmond SJ, et al. Clinicopathological correlations in corticobasal degeneration. Ann Neurol. 2011;70:327–40.PubMedPubMedCentralCrossRef
69.
70.
go back to reference Vaillancourt DE, Prodoehl J, Abraham I, Corcos DM, Zhou XJ, Cornelia CL, et al. High-resolution diffusion tensor imaging in the substantia nigra of de novo Parkinson disease. Neurology. 2009;72:1378–84.PubMedPubMedCentralCrossRef Vaillancourt DE, Prodoehl J, Abraham I, Corcos DM, Zhou XJ, Cornelia CL, et al. High-resolution diffusion tensor imaging in the substantia nigra of de novo Parkinson disease. Neurology. 2009;72:1378–84.PubMedPubMedCentralCrossRef
71.
go back to reference Scherfler C, Schocke MF, Seppi K, Esterhammer R, Brenneis C, Jaschke W, et al. Voxel-wise analysis of diffusion weighted imaging reveals disruption of the olfactory tract in Parkinson’s disease. Brain. 2006;129:538–42.PubMedCrossRef Scherfler C, Schocke MF, Seppi K, Esterhammer R, Brenneis C, Jaschke W, et al. Voxel-wise analysis of diffusion weighted imaging reveals disruption of the olfactory tract in Parkinson’s disease. Brain. 2006;129:538–42.PubMedCrossRef
72.
go back to reference Rolheiser TM, Fulton HG, Good KP, Fisk JD, McKelvey JR, Scherfler C, et al. Diffusion tensor imaging and olfactory identification testing in early-stage Parkinson’s disease. J Neurol. 2011;258:1254–60.PubMedCrossRef Rolheiser TM, Fulton HG, Good KP, Fisk JD, McKelvey JR, Scherfler C, et al. Diffusion tensor imaging and olfactory identification testing in early-stage Parkinson’s disease. J Neurol. 2011;258:1254–60.PubMedCrossRef
73.
go back to reference Boelmans K, Bodammer NC, Suchorska B, Kaufmann J, Ebersbach G, Heinze HJ, et al. Diffusion tensor imaging of the corpus callosum differentiates corticobasal syndrome from Parkinson’s disease. Park Relat Disord. 2010;16:498–502.CrossRef Boelmans K, Bodammer NC, Suchorska B, Kaufmann J, Ebersbach G, Heinze HJ, et al. Diffusion tensor imaging of the corpus callosum differentiates corticobasal syndrome from Parkinson’s disease. Park Relat Disord. 2010;16:498–502.CrossRef
74.
go back to reference Meijer FJA, van Rumund A, Tuladhar AM, Aerts MB, Titulaer I, Esselink RAJ, et al. Conventional 3 T brain MRI and diffusion tensor imaging in the diagnostic workup of early stage parkinsonism. Neuroradiology. 2015;57:655–69.PubMedPubMedCentralCrossRef Meijer FJA, van Rumund A, Tuladhar AM, Aerts MB, Titulaer I, Esselink RAJ, et al. Conventional 3 T brain MRI and diffusion tensor imaging in the diagnostic workup of early stage parkinsonism. Neuroradiology. 2015;57:655–69.PubMedPubMedCentralCrossRef
75.
go back to reference Barbagallo G, Sierra-Peña M, Nemmi F, Le Traon AP, Meissner WG, Rascol O, et al. Multimodal MRI assessment of nigro-striatal pathway in multiple system atrophy and Parkinson disease. Mov Disord. 2016;31:325–34.PubMedCrossRef Barbagallo G, Sierra-Peña M, Nemmi F, Le Traon AP, Meissner WG, Rascol O, et al. Multimodal MRI assessment of nigro-striatal pathway in multiple system atrophy and Parkinson disease. Mov Disord. 2016;31:325–34.PubMedCrossRef
76.
go back to reference Ito M, Watanabe H, Kawai Y, Atsuta N, Tanaka F, Naganawa S, et al. Usefulness of combined fractional anisotropy and apparent diffusion coefficient values for detection of involvement in multiple system atrophy. J Neurol Neurosurg Psychiatry. 2007;78:722–8.PubMedPubMedCentralCrossRef Ito M, Watanabe H, Kawai Y, Atsuta N, Tanaka F, Naganawa S, et al. Usefulness of combined fractional anisotropy and apparent diffusion coefficient values for detection of involvement in multiple system atrophy. J Neurol Neurosurg Psychiatry. 2007;78:722–8.PubMedPubMedCentralCrossRef
77.
go back to reference Shiga K, Yamada K, Yoshikawa K, Mizuno T, Nishimura T, Nakagawa M. Local tissue anisotropy decreases in cerebellopetal fibers and pyramidal tract in multiple system atrophy. J Neurol. 2005;252:589–96.PubMedCrossRef Shiga K, Yamada K, Yoshikawa K, Mizuno T, Nishimura T, Nakagawa M. Local tissue anisotropy decreases in cerebellopetal fibers and pyramidal tract in multiple system atrophy. J Neurol. 2005;252:589–96.PubMedCrossRef
78.
go back to reference Blain CRV, Barker GJ, Jarosz JM, Coyle NA, Landau S, Brown RG, et al. Measuring brain stem and cerebellar damage in parkinsonian syndromes using diffusion tensor MRI. Neurology. 2006;67:2199–205.PubMedCrossRef Blain CRV, Barker GJ, Jarosz JM, Coyle NA, Landau S, Brown RG, et al. Measuring brain stem and cerebellar damage in parkinsonian syndromes using diffusion tensor MRI. Neurology. 2006;67:2199–205.PubMedCrossRef
79.
go back to reference Oishi K, Konishi J, Mori S, Ishihara H, Kawamitsu H, Fujii M, et al. Reduced fractional anisotropy in early-stage cerebellar variant of multiple system atrophy. J Neuroimaging. 2009;19:127–31.PubMedCrossRef Oishi K, Konishi J, Mori S, Ishihara H, Kawamitsu H, Fujii M, et al. Reduced fractional anisotropy in early-stage cerebellar variant of multiple system atrophy. J Neuroimaging. 2009;19:127–31.PubMedCrossRef
80.
go back to reference Pellecchia MT, Barone P, Mollica C, Salvatore E, Ianniciello M, Longo K, et al. Diffusion-weighted imaging in multiple system atrophy: a comparison between clinical subtypes. Mov Disord. 2009;24:689–96.PubMedCrossRef Pellecchia MT, Barone P, Mollica C, Salvatore E, Ianniciello M, Longo K, et al. Diffusion-weighted imaging in multiple system atrophy: a comparison between clinical subtypes. Mov Disord. 2009;24:689–96.PubMedCrossRef
81.
go back to reference Erbetta A, Mandelli ML, Savoiardo M, Grisoli M, Bizzi A, Soliveri P, et al. Diffusion tensor imaging shows different topographic involvement of the thalamus in progressive supranuclear palsy and corticobasal degeneration. Am J Neuroradiol. 2009;30:1482–7.PubMedCrossRef Erbetta A, Mandelli ML, Savoiardo M, Grisoli M, Bizzi A, Soliveri P, et al. Diffusion tensor imaging shows different topographic involvement of the thalamus in progressive supranuclear palsy and corticobasal degeneration. Am J Neuroradiol. 2009;30:1482–7.PubMedCrossRef
82.
go back to reference Kvickström P, Eriksson B, van Westen D, Lätt J, Elfgren C, Nilsson C. Selective frontal neurodegeneration of the inferior fronto-occipital fasciculus in progressive supranuclear palsy (PSP) demonstrated by diffusion tensor tractography. BMC Neurol. 2011;11:13.PubMedPubMedCentralCrossRef Kvickström P, Eriksson B, van Westen D, Lätt J, Elfgren C, Nilsson C. Selective frontal neurodegeneration of the inferior fronto-occipital fasciculus in progressive supranuclear palsy (PSP) demonstrated by diffusion tensor tractography. BMC Neurol. 2011;11:13.PubMedPubMedCentralCrossRef
83.
go back to reference Seppi K, Schocke MFH, Esterhammer R, Kremser C, Brenneis C, Mueller J, et al. Diffusion-weighted imaging discriminates progressive supranuclear palsy from PD, but not from the parkinson variant of multiple system atrophy. Neurology. 2003;60:922–7.PubMedCrossRef Seppi K, Schocke MFH, Esterhammer R, Kremser C, Brenneis C, Mueller J, et al. Diffusion-weighted imaging discriminates progressive supranuclear palsy from PD, but not from the parkinson variant of multiple system atrophy. Neurology. 2003;60:922–7.PubMedCrossRef
84.
go back to reference Bozzali M, Falini A, Cercignani M, Baglio F, Farina E, Alberoni M, et al. Brain tissue damage in dementia with Lewy bodies: an in vivo diffusion tensor MRI study. Brain. 2005;128:1595–604.PubMedCrossRef Bozzali M, Falini A, Cercignani M, Baglio F, Farina E, Alberoni M, et al. Brain tissue damage in dementia with Lewy bodies: an in vivo diffusion tensor MRI study. Brain. 2005;128:1595–604.PubMedCrossRef
85.
go back to reference Kantarci K, Avula R, Senjem ML, Samikoglu AR, Zhang B, Weigand SD, et al. Dementia with Lewy bodies and Alzheimer disease: neurodegenerative patterns characterized by DTI. Neurology. 2010;74:1814–21.PubMedPubMedCentralCrossRef Kantarci K, Avula R, Senjem ML, Samikoglu AR, Zhang B, Weigand SD, et al. Dementia with Lewy bodies and Alzheimer disease: neurodegenerative patterns characterized by DTI. Neurology. 2010;74:1814–21.PubMedPubMedCentralCrossRef
86.
go back to reference Firbank MJ, Blamire AM, Krishnan MS, Teodorczuk A, English P, Gholkar A, et al. Diffusion tensor imaging in dementia with Lewy bodies and Alzheimer’s disease. Psychiatry Res. 2007;155:135–45.PubMedCrossRef Firbank MJ, Blamire AM, Krishnan MS, Teodorczuk A, English P, Gholkar A, et al. Diffusion tensor imaging in dementia with Lewy bodies and Alzheimer’s disease. Psychiatry Res. 2007;155:135–45.PubMedCrossRef
87.
go back to reference Matsui H, Nishinaka K, Oda M, Niikawa H, Kubori T, Udaka F. Dementia in Parkinson’s disease: diffusion tensor imaging. Acta Neurol Scand. 2007;116:177–81.PubMedCrossRef Matsui H, Nishinaka K, Oda M, Niikawa H, Kubori T, Udaka F. Dementia in Parkinson’s disease: diffusion tensor imaging. Acta Neurol Scand. 2007;116:177–81.PubMedCrossRef
88.
go back to reference Firbank MJ, Harrison RM, O’Brien JT. A comprehensive review of proton magnetic resonance spectroscopy studies in dementia and Parkinson’s disease. Dement Geriatr Cogn Disord. 2002;14:64–76.PubMedCrossRef Firbank MJ, Harrison RM, O’Brien JT. A comprehensive review of proton magnetic resonance spectroscopy studies in dementia and Parkinson’s disease. Dement Geriatr Cogn Disord. 2002;14:64–76.PubMedCrossRef
89.
go back to reference Taylor-Robinson SD, Turjanski N, Bhattacharya S, Seery JP, Sargentoni J, Brooks DJ, et al. A proton magnetic resonance spectroscopy study of the striatum and cerebral cortex in Parkinson’s disease. Metab Brain Dis. 1999;14:45–55.PubMedCrossRef Taylor-Robinson SD, Turjanski N, Bhattacharya S, Seery JP, Sargentoni J, Brooks DJ, et al. A proton magnetic resonance spectroscopy study of the striatum and cerebral cortex in Parkinson’s disease. Metab Brain Dis. 1999;14:45–55.PubMedCrossRef
90.
go back to reference Camicioli RM, Hanstock CC, Bouchard TP, Gee M, Fisher NJ, Martin WRW. Magnetic resonance spectroscopic evidence for presupplementary motor area neuronal dysfunction in Parkinson’s disease. Mov Disord. 2007;22:382–6.PubMedCrossRef Camicioli RM, Hanstock CC, Bouchard TP, Gee M, Fisher NJ, Martin WRW. Magnetic resonance spectroscopic evidence for presupplementary motor area neuronal dysfunction in Parkinson’s disease. Mov Disord. 2007;22:382–6.PubMedCrossRef
91.
go back to reference Camicioli RM, Korzan JR, Foster SL, Fisher NJ, Emery DJ, Bastos AC, et al. Posterior cingulate metabolic changes occur in Parkinson’s disease patients without dementia. Neurosci Lett. 2004;354:177–80.PubMedCrossRef Camicioli RM, Korzan JR, Foster SL, Fisher NJ, Emery DJ, Bastos AC, et al. Posterior cingulate metabolic changes occur in Parkinson’s disease patients without dementia. Neurosci Lett. 2004;354:177–80.PubMedCrossRef
92.
go back to reference Gröger A, Bender B, Wurster I, Chadzynski GL, Klose U, Berg D. Differentiation between idiopathic and atypical Parkinsonian syndromes using three-dimensional magnetic resonance spectroscopic imaging. J Neurol Neurosurg Psychiatry. 2013;84:644–9.PubMedCrossRef Gröger A, Bender B, Wurster I, Chadzynski GL, Klose U, Berg D. Differentiation between idiopathic and atypical Parkinsonian syndromes using three-dimensional magnetic resonance spectroscopic imaging. J Neurol Neurosurg Psychiatry. 2013;84:644–9.PubMedCrossRef
93.
go back to reference Federico F, Simone IL, Lucivero V, De Mari M, Giannini P, Iliceto G, et al. Proton magnetic resonance spectroscopy in Parkinson’s disease and progressive Supranuclear palsy. J Neurol Neurosurg Psychiatry. 1997;62:239–42.PubMedPubMedCentralCrossRef Federico F, Simone IL, Lucivero V, De Mari M, Giannini P, Iliceto G, et al. Proton magnetic resonance spectroscopy in Parkinson’s disease and progressive Supranuclear palsy. J Neurol Neurosurg Psychiatry. 1997;62:239–42.PubMedPubMedCentralCrossRef
94.
go back to reference Negoro K, Tada Y, Ogasawara J, Kawai M, Morimatsu M, Hashida M, et al. Proton magnetic resonance spectroscopy in corticobasal degeneration and progressive supranuclear palsy. Geriatr Gerontol Int. 2004;4:84–92.CrossRef Negoro K, Tada Y, Ogasawara J, Kawai M, Morimatsu M, Hashida M, et al. Proton magnetic resonance spectroscopy in corticobasal degeneration and progressive supranuclear palsy. Geriatr Gerontol Int. 2004;4:84–92.CrossRef
95.
go back to reference Abe K, Terakawa H, Takanashi M, Watanabe Y, Tanaka H, Fujita N, et al. Proton magnetic resonance spectroscopy of patients with parkinsonism. Brain Res Bull. 2000;52:589–95.PubMedCrossRef Abe K, Terakawa H, Takanashi M, Watanabe Y, Tanaka H, Fujita N, et al. Proton magnetic resonance spectroscopy of patients with parkinsonism. Brain Res Bull. 2000;52:589–95.PubMedCrossRef
96.
go back to reference Watanabe H, Fukatsu H, Katsuno M, Sugiura M, Hamada K, Okada Y, et al. Multiple regional 1H-MR spectroscopy in multiple system atrophy: NAA/Cr reduction in pontine base as a valuable diagnostic marker. J Neurol Neurosurg Psychiatry. 2004;75:103–9.PubMedPubMedCentral Watanabe H, Fukatsu H, Katsuno M, Sugiura M, Hamada K, Okada Y, et al. Multiple regional 1H-MR spectroscopy in multiple system atrophy: NAA/Cr reduction in pontine base as a valuable diagnostic marker. J Neurol Neurosurg Psychiatry. 2004;75:103–9.PubMedPubMedCentral
97.
go back to reference Tedeschi G, Litvan I, Bonavita S, Bertolino A, Lundbom N, Patronas NJ, et al. Proton magnetic resonance spectroscopic imaging in progressive supranuclear palsy, Parkinson’s disease and corticobasal degeneration. Brain. 1997;120:1541–52.PubMedCrossRef Tedeschi G, Litvan I, Bonavita S, Bertolino A, Lundbom N, Patronas NJ, et al. Proton magnetic resonance spectroscopic imaging in progressive supranuclear palsy, Parkinson’s disease and corticobasal degeneration. Brain. 1997;120:1541–52.PubMedCrossRef
98.
go back to reference Griffith HR, den Hollander JA, Okonkwo OC, O’Brien T, Watts RL, Marson DC. Brain N-acetylaspartate is reduced in Parkinson disease with dementia. Alzheimer Dis Assoc Disord. 2008;22:54–60.PubMedCrossRef Griffith HR, den Hollander JA, Okonkwo OC, O’Brien T, Watts RL, Marson DC. Brain N-acetylaspartate is reduced in Parkinson disease with dementia. Alzheimer Dis Assoc Disord. 2008;22:54–60.PubMedCrossRef
99.
go back to reference Xuan X, Ding M, Gong X. Proton magnetic resonance spectroscopy detects a relative decrease of N-acetylaspartate in the hippocampus of patients with dementia with Lewy bodies. J Neuroimaging. 2008;18:137–41.PubMedCrossRef Xuan X, Ding M, Gong X. Proton magnetic resonance spectroscopy detects a relative decrease of N-acetylaspartate in the hippocampus of patients with dementia with Lewy bodies. J Neuroimaging. 2008;18:137–41.PubMedCrossRef
100.
go back to reference Mazuel L, Chassain C, Jean B, Pereira B, Cladiere A, Speziale C, et al. Proton MR spectroscopy for diagnosis and evaluation of treatment efficacy in Parkinson disease. Radiology. 2015;278:142764. Mazuel L, Chassain C, Jean B, Pereira B, Cladiere A, Speziale C, et al. Proton MR spectroscopy for diagnosis and evaluation of treatment efficacy in Parkinson disease. Radiology. 2015;278:142764.
101.
go back to reference Beaulieu J-M, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217.PubMedCrossRef Beaulieu J-M, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217.PubMedCrossRef
102.
go back to reference Djang DSW, Janssen MJR, Bohnen N, Booij J, Henderson TA, Herholz K, et al. SNM practice guideline for dopamine transporter imaging with 123I-ioflupane SPECT 1.0. J Nucl Med. 2012;53:154–63.PubMedCrossRef Djang DSW, Janssen MJR, Bohnen N, Booij J, Henderson TA, Herholz K, et al. SNM practice guideline for dopamine transporter imaging with 123I-ioflupane SPECT 1.0. J Nucl Med. 2012;53:154–63.PubMedCrossRef
103.
go back to reference Catafau AM, Tolosa E, Laloux P, Vander Borght T, Van Zandijcke M, De Geeter F, et al. Impact of dopamine transporter SPECT using 123I-Ioflupane on diagnosis and management of patients with clinically uncertain parkinsonian syndromes. Mov Disord. 2004;19:1175–82.PubMedCrossRef Catafau AM, Tolosa E, Laloux P, Vander Borght T, Van Zandijcke M, De Geeter F, et al. Impact of dopamine transporter SPECT using 123I-Ioflupane on diagnosis and management of patients with clinically uncertain parkinsonian syndromes. Mov Disord. 2004;19:1175–82.PubMedCrossRef
104.
go back to reference Brücke T, Asenbaum S, Pirker W, Djamshidian S, Wenger S, Wöber C, et al. Measurement of the dopaminergic degeneration in Parkinson’s disease with [123I] beta-CIT and SPECT. Correlation with clinical findings and comparison with multiple system atrophy and progressive Supranuclear palsy. J Neural Transm Suppl. 1997;50:9–24.PubMedCrossRef Brücke T, Asenbaum S, Pirker W, Djamshidian S, Wenger S, Wöber C, et al. Measurement of the dopaminergic degeneration in Parkinson’s disease with [123I] beta-CIT and SPECT. Correlation with clinical findings and comparison with multiple system atrophy and progressive Supranuclear palsy. J Neural Transm Suppl. 1997;50:9–24.PubMedCrossRef
105.
go back to reference Plotkin M, Amthauer H, Klaffke S, Kühn A, Lüdemann L, Arnold G, et al. Combined 123I-FP-CIT and 123I-IBZM SPECT for the diagnosis of parkinsonian syndromes: Study on 72 patients. J Neural Transm. 2005;112:677–92.PubMedCrossRef Plotkin M, Amthauer H, Klaffke S, Kühn A, Lüdemann L, Arnold G, et al. Combined 123I-FP-CIT and 123I-IBZM SPECT for the diagnosis of parkinsonian syndromes: Study on 72 patients. J Neural Transm. 2005;112:677–92.PubMedCrossRef
106.
go back to reference Politis M. Neuroimaging in Parkinson disease: from research setting to clinical practice. Nat Rev Neurol. 2014;10:708–22.PubMedCrossRef Politis M. Neuroimaging in Parkinson disease: from research setting to clinical practice. Nat Rev Neurol. 2014;10:708–22.PubMedCrossRef
107.
go back to reference Im J-H, Chung SJ, Kim J-S, Lee MC. Differential patterns of dopamine transporter loss in the basal ganglia of progressive supranuclear palsy and Parkinson’s disease: analysis with [(123)I]IPT single photon emission computed tomography. J Neurol Sci. 2006;244:103–9.PubMedCrossRef Im J-H, Chung SJ, Kim J-S, Lee MC. Differential patterns of dopamine transporter loss in the basal ganglia of progressive supranuclear palsy and Parkinson’s disease: analysis with [(123)I]IPT single photon emission computed tomography. J Neurol Sci. 2006;244:103–9.PubMedCrossRef
108.
go back to reference Klaffke S, Kuhn AA, Plotkin M, Amthauer H, Harnack D, Felix R, et al. Dopamine transporters, D2 receptors, and glucose metabolism in corticobasal degeneration. Mov Disord. 2006;21:1724–7.PubMedCrossRef Klaffke S, Kuhn AA, Plotkin M, Amthauer H, Harnack D, Felix R, et al. Dopamine transporters, D2 receptors, and glucose metabolism in corticobasal degeneration. Mov Disord. 2006;21:1724–7.PubMedCrossRef
109.
go back to reference O’Brien JT, Colloby S, Fenwick J, Williams ED, Firbank M, Burn D, et al. Dopamine transporter loss visualized with FP-CIT SPECT in the differential diagnosis of dementia with Lewy bodies. Arch Neurol. 2004;61:919–25.PubMedCrossRef O’Brien JT, Colloby S, Fenwick J, Williams ED, Firbank M, Burn D, et al. Dopamine transporter loss visualized with FP-CIT SPECT in the differential diagnosis of dementia with Lewy bodies. Arch Neurol. 2004;61:919–25.PubMedCrossRef
110.
go back to reference Walker Z, Costa DC, Walker RWH, Shaw K, Gacinovic S, Stevens T, et al. Differentiation of dementia with Lewy bodies from Alzheimer’s disease using a dopaminergic presynaptic ligand. J Neurol Neurosurg Psychiatry. 2002;73:134–40.PubMedPubMedCentralCrossRef Walker Z, Costa DC, Walker RWH, Shaw K, Gacinovic S, Stevens T, et al. Differentiation of dementia with Lewy bodies from Alzheimer’s disease using a dopaminergic presynaptic ligand. J Neurol Neurosurg Psychiatry. 2002;73:134–40.PubMedPubMedCentralCrossRef
111.
go back to reference Kim YJ, Ichise M, Ballinger JR, Vines D, Erami SS, Tatschida T, et al. Combination of dopamine transporter and D2 receptor SPECT in the diagnostic evaluation of PD, MSA, and PSP. Mov Disord. 2002;303–12. Kim YJ, Ichise M, Ballinger JR, Vines D, Erami SS, Tatschida T, et al. Combination of dopamine transporter and D2 receptor SPECT in the diagnostic evaluation of PD, MSA, and PSP. Mov Disord. 2002;303–12.
112.
go back to reference Del Sole A, Perini G, Lecchi M, Mariani C, Lucignani G, Clerici F. Correlation between 123I-FP-CIT brain SPECT and parkinsonism in dementia with Lewy bodies: caveat for clinical use. Clin Nucl Med. 2015;40:32–5.PubMedCrossRef Del Sole A, Perini G, Lecchi M, Mariani C, Lucignani G, Clerici F. Correlation between 123I-FP-CIT brain SPECT and parkinsonism in dementia with Lewy bodies: caveat for clinical use. Clin Nucl Med. 2015;40:32–5.PubMedCrossRef
113.
go back to reference Spiegel J, Möllers MO, Jost WH, Fuss G, Samnick S, Dillmann U, et al. FP-CIT and MIBG scintigraphy in early Parkinson’s disease. Mov Disord. 2005;20:552–61.PubMedCrossRef Spiegel J, Möllers MO, Jost WH, Fuss G, Samnick S, Dillmann U, et al. FP-CIT and MIBG scintigraphy in early Parkinson’s disease. Mov Disord. 2005;20:552–61.PubMedCrossRef
114.
go back to reference Benamer HTS, Patterson J, Wyper DJ, Hadley DM, Macphee GJA, Grosset DG. Correlation of Parkinson’s disease severity and duration with 123I-FP-CIT SPECT Striatal uptake. Mov Disord. 2000;15:692–8.PubMedCrossRef Benamer HTS, Patterson J, Wyper DJ, Hadley DM, Macphee GJA, Grosset DG. Correlation of Parkinson’s disease severity and duration with 123I-FP-CIT SPECT Striatal uptake. Mov Disord. 2000;15:692–8.PubMedCrossRef
115.
go back to reference Vlaar AMM, van Kroonenburgh MJPG, Kessels AGH, Weber WEJ. Meta-analysis of the literature on diagnostic accuracy of SPECT in Parkinsonian syndromes. BMC Neurol. 2007;7:27.PubMedPubMedCentralCrossRef Vlaar AMM, van Kroonenburgh MJPG, Kessels AGH, Weber WEJ. Meta-analysis of the literature on diagnostic accuracy of SPECT in Parkinsonian syndromes. BMC Neurol. 2007;7:27.PubMedPubMedCentralCrossRef
116.
go back to reference Benamer HTS, Patterson J, Grosset DG, Booij J, De Bruin K, Van Royen E, et al. Accurate differentiation of parkinsonism and essential tremor using visual assessment of [123I]-FP-CIT SPECT imaging: The [123I]-FP-CIT study group. Mov Disord. 2000;15:503–10.PubMedCrossRef Benamer HTS, Patterson J, Grosset DG, Booij J, De Bruin K, Van Royen E, et al. Accurate differentiation of parkinsonism and essential tremor using visual assessment of [123I]-FP-CIT SPECT imaging: The [123I]-FP-CIT study group. Mov Disord. 2000;15:503–10.PubMedCrossRef
117.
go back to reference Nocker M, Seppi K, Donnemiller E, Virgolini I, Wenning GK, Poewe W, et al. Progression of dopamine transporter decline in patients with the Parkinson variant of multiple system atrophy: a voxel-based analysis of [123I]β-CIT SPECT. Eur J Nucl Med Mol Imaging. 2012;39:1012–20.PubMedCrossRef Nocker M, Seppi K, Donnemiller E, Virgolini I, Wenning GK, Poewe W, et al. Progression of dopamine transporter decline in patients with the Parkinson variant of multiple system atrophy: a voxel-based analysis of [123I]β-CIT SPECT. Eur J Nucl Med Mol Imaging. 2012;39:1012–20.PubMedCrossRef
118.
go back to reference Antonini A, Benti R, De Notaris R, Tesei S, Zecchinelli A, Sacilotto G, et al. 123I-Ioflupane/SPECT binding to striatal dopamine transporter (DAT) uptake in patients with Parkinson’s disease, multiple system atrophy, and progressive supranuclear palsy. Neurol Sci. 2003;24:149–50.PubMedCrossRef Antonini A, Benti R, De Notaris R, Tesei S, Zecchinelli A, Sacilotto G, et al. 123I-Ioflupane/SPECT binding to striatal dopamine transporter (DAT) uptake in patients with Parkinson’s disease, multiple system atrophy, and progressive supranuclear palsy. Neurol Sci. 2003;24:149–50.PubMedCrossRef
119.
go back to reference Filippi L, Manni C, Pierantozzi M, Brusa L, Danieli R, Stanzione P, et al. 123I-FP-CIT in progressive supranuclear palsy and in Parkinson’s disease: a SPECT semiquantitative study. Nucl Med Commun. 2006;27:381–6.PubMedCrossRef Filippi L, Manni C, Pierantozzi M, Brusa L, Danieli R, Stanzione P, et al. 123I-FP-CIT in progressive supranuclear palsy and in Parkinson’s disease: a SPECT semiquantitative study. Nucl Med Commun. 2006;27:381–6.PubMedCrossRef
120.
go back to reference O’Brien JT, McKeith IG, Walker Z, Tatsch K, Booij J, Darcourt J, et al. Diagnostic accuracy of 123I-FP-CIT SPECT in possible dementia with Lewy bodies. Br J Psychiatry. 2009;194:34–9.PubMedCrossRef O’Brien JT, McKeith IG, Walker Z, Tatsch K, Booij J, Darcourt J, et al. Diagnostic accuracy of 123I-FP-CIT SPECT in possible dementia with Lewy bodies. Br J Psychiatry. 2009;194:34–9.PubMedCrossRef
121.
go back to reference Walker Z, Jaros E, Walker RWH, Lee L, Costa DC, Livingston G, et al. Dementia with Lewy bodies: a comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry. 2007;78:1176–81.PubMedPubMedCentralCrossRef Walker Z, Jaros E, Walker RWH, Lee L, Costa DC, Livingston G, et al. Dementia with Lewy bodies: a comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry. 2007;78:1176–81.PubMedPubMedCentralCrossRef
122.
go back to reference Brigo F, Turri G, Tinazzi M. 123I-FP-CIT SPECT in the differential diagnosis between dementia with Lewy bodies and other dementias. J Neurol Sci. 2015;359:161–71.PubMedCrossRef Brigo F, Turri G, Tinazzi M. 123I-FP-CIT SPECT in the differential diagnosis between dementia with Lewy bodies and other dementias. J Neurol Sci. 2015;359:161–71.PubMedCrossRef
123.
go back to reference Papathanasiou ND, Boutsiadis A, Dickson J, Bomanji JB. Diagnostic accuracy of 123I-FP-CIT (DaTSCAN) in dementia with Lewy bodies: a meta-analysis of published studies. Parkinsonism Relat Disord. 2012;18:225–9.PubMedCrossRef Papathanasiou ND, Boutsiadis A, Dickson J, Bomanji JB. Diagnostic accuracy of 123I-FP-CIT (DaTSCAN) in dementia with Lewy bodies: a meta-analysis of published studies. Parkinsonism Relat Disord. 2012;18:225–9.PubMedCrossRef
124.
go back to reference Cilia R, Rossi C, Frosini D, Volterrani D, Siri C, Pagni C, et al. Dopamine transporter spect imaging in corticobasal syndrome. PLoS ONE. 2011;6:e18301.PubMedPubMedCentralCrossRef Cilia R, Rossi C, Frosini D, Volterrani D, Siri C, Pagni C, et al. Dopamine transporter spect imaging in corticobasal syndrome. PLoS ONE. 2011;6:e18301.PubMedPubMedCentralCrossRef
125.
go back to reference Pirker S, Perju-Dumbrava L, Kovacs GG, Traub-Weidinger T, Pirker W. Progressive dopamine transporter binding loss in autopsy-confirmed corticobasal degeneration. J Parkinsons Dis. 2015;5:907–12.PubMedCrossRef Pirker S, Perju-Dumbrava L, Kovacs GG, Traub-Weidinger T, Pirker W. Progressive dopamine transporter binding loss in autopsy-confirmed corticobasal degeneration. J Parkinsons Dis. 2015;5:907–12.PubMedCrossRef
126.
go back to reference Erro R, Schneider SA, Quinn NP, Bhatia KP. What do patients with scans without evidence of dopaminergic deficit (SWEDD) have? New evidence and continuing controversies. J Neurol Neurosurg Psychiatry. 2016:87(3):319-23. Erro R, Schneider SA, Quinn NP, Bhatia KP. What do patients with scans without evidence of dopaminergic deficit (SWEDD) have? New evidence and continuing controversies. J Neurol Neurosurg Psychiatry. 2016:87(3):319-23.
127.
go back to reference Schneider SA, Edwards MJ, Mir P, Cordivari C, Hooker J, Dickson J, et al. Patients with adult-onset dystonic tremor resembling Parkinsonian tremor have scans without evidence of dopaminergic deficit (SWEDDs). Mov Disord. 2007;22:2210–5.PubMedCrossRef Schneider SA, Edwards MJ, Mir P, Cordivari C, Hooker J, Dickson J, et al. Patients with adult-onset dystonic tremor resembling Parkinsonian tremor have scans without evidence of dopaminergic deficit (SWEDDs). Mov Disord. 2007;22:2210–5.PubMedCrossRef
128.
go back to reference Marshall VL, Patterson J, Hadley DM, Grosset KA, Grosset DG. Two-year follow-up in 150 consecutive cases with normal dopamine transporter imaging. Nucl Med Commun. 2006;27:933–7.PubMedCrossRef Marshall VL, Patterson J, Hadley DM, Grosset KA, Grosset DG. Two-year follow-up in 150 consecutive cases with normal dopamine transporter imaging. Nucl Med Commun. 2006;27:933–7.PubMedCrossRef
129.
go back to reference van Royen E, Verhoeff NF, Speelman JD, Wolters EC, Kuiper MA, Janssen AG. Multiple system atrophy and progressive Supranuclear palsy. Diminished Striatal D2 dopamine receptor activity demonstrated by 123I-IBZM single photon emission computed tomography. Arch Neurol. 1993;50:513–6.PubMedCrossRef van Royen E, Verhoeff NF, Speelman JD, Wolters EC, Kuiper MA, Janssen AG. Multiple system atrophy and progressive Supranuclear palsy. Diminished Striatal D2 dopamine receptor activity demonstrated by 123I-IBZM single photon emission computed tomography. Arch Neurol. 1993;50:513–6.PubMedCrossRef
130.
go back to reference Ichise M, Kim YJ, Ballinger JR, Vines D, Erami SS, Tanaka F, et al. SPECT imaging of pre- and postsynaptic dopaminergic alterations in L-dopa-untreated PD. Neurology. 1999;52:1206–14.PubMedCrossRef Ichise M, Kim YJ, Ballinger JR, Vines D, Erami SS, Tanaka F, et al. SPECT imaging of pre- and postsynaptic dopaminergic alterations in L-dopa-untreated PD. Neurology. 1999;52:1206–14.PubMedCrossRef
131.
go back to reference Pirker S, Perju-Dumbrava L, Kovacs GG, Traub-Weidinger T, Asenbaum S, Pirker W. Dopamine D2 receptor SPECT in corticobasal syndrome and autopsy-confirmed corticobasal degeneration. Park Relat Disord. 2013;19:222–6.CrossRef Pirker S, Perju-Dumbrava L, Kovacs GG, Traub-Weidinger T, Asenbaum S, Pirker W. Dopamine D2 receptor SPECT in corticobasal syndrome and autopsy-confirmed corticobasal degeneration. Park Relat Disord. 2013;19:222–6.CrossRef
132.
go back to reference Koch W, Hamann C, Radau PE, Tatsch K. Does combined imaging of the pre- and postsynaptic dopaminergic system increase the diagnostic accuracy in the differential diagnosis of parkinsonism? Eur J Nucl Med Mol Imaging. 2007;34:1265–73.PubMedCrossRef Koch W, Hamann C, Radau PE, Tatsch K. Does combined imaging of the pre- and postsynaptic dopaminergic system increase the diagnostic accuracy in the differential diagnosis of parkinsonism? Eur J Nucl Med Mol Imaging. 2007;34:1265–73.PubMedCrossRef
133.
go back to reference Lobotesis K, Fenwick JD, Phipps A, Ryman A, Swann A, Ballard C, et al. Occipital hypoperfusion on SPECT in dementia with Lewy bodies but not AD. Neurology. 2001;56:643–9.PubMedCrossRef Lobotesis K, Fenwick JD, Phipps A, Ryman A, Swann A, Ballard C, et al. Occipital hypoperfusion on SPECT in dementia with Lewy bodies but not AD. Neurology. 2001;56:643–9.PubMedCrossRef
134.
go back to reference Song IU, Yoo I, Chung YA, Jeong J. The value of brain perfusion SPECT for differentiation between mildly symptomatic idiopathic Parkinson’s disease and the Parkinson variant of multiple system atrophy. Nucl Med Commun England. 2015;36:1049–54.CrossRef Song IU, Yoo I, Chung YA, Jeong J. The value of brain perfusion SPECT for differentiation between mildly symptomatic idiopathic Parkinson’s disease and the Parkinson variant of multiple system atrophy. Nucl Med Commun England. 2015;36:1049–54.CrossRef
135.
go back to reference Antonini A, De Notaris R, Benti R, De Gaspari D, Pezzoli G. Perfusion ECD/SPECT in the characterization of cognitive deficits in Parkinson’s disease. Neurol Sci. 2001;22:45–6.PubMedCrossRef Antonini A, De Notaris R, Benti R, De Gaspari D, Pezzoli G. Perfusion ECD/SPECT in the characterization of cognitive deficits in Parkinson’s disease. Neurol Sci. 2001;22:45–6.PubMedCrossRef
136.
go back to reference Matsui H, Udaka F, Miyoshi T, Hara N, Tamura A, Oda M, et al. Brain perfusion differences between Parkinson’s disease and multiple system atrophy with predominant parkinsonian features. Park Relat Disord. 2005;11:227–32.CrossRef Matsui H, Udaka F, Miyoshi T, Hara N, Tamura A, Oda M, et al. Brain perfusion differences between Parkinson’s disease and multiple system atrophy with predominant parkinsonian features. Park Relat Disord. 2005;11:227–32.CrossRef
137.
go back to reference Firbank MJ, Molloy S, McKeith IG, Burn DJ, O’Brien JT. Longitudinal change in 99mTcHMPAO cerebral perfusion SPECT in Parkinson’s disease over one year. J Neurol Neurosurg Psychiatry. 2005;76:1448–51.PubMedPubMedCentralCrossRef Firbank MJ, Molloy S, McKeith IG, Burn DJ, O’Brien JT. Longitudinal change in 99mTcHMPAO cerebral perfusion SPECT in Parkinson’s disease over one year. J Neurol Neurosurg Psychiatry. 2005;76:1448–51.PubMedPubMedCentralCrossRef
138.
go back to reference Matsuda H, Imabayashi E, Kuji I, Seto A, Ito K, Kikuta D, et al. Evaluation of both perfusion and atrophy in multiple system atrophy of the cerebellar type using brain SPECT alone. BMC Med Imaging. 2010;10:17.PubMedPubMedCentralCrossRef Matsuda H, Imabayashi E, Kuji I, Seto A, Ito K, Kikuta D, et al. Evaluation of both perfusion and atrophy in multiple system atrophy of the cerebellar type using brain SPECT alone. BMC Med Imaging. 2010;10:17.PubMedPubMedCentralCrossRef
139.
go back to reference Nurmi E, Ruottinen HM, Bergman J, Haaparanta M, Solin O, Sonninen P, et al. Rate of progression in Parkinson’s disease: A 6-[18 F]fluoro-L-dopa PET study. Mov Disord. 2001;16:608–15.PubMedCrossRef Nurmi E, Ruottinen HM, Bergman J, Haaparanta M, Solin O, Sonninen P, et al. Rate of progression in Parkinson’s disease: A 6-[18 F]fluoro-L-dopa PET study. Mov Disord. 2001;16:608–15.PubMedCrossRef
140.
go back to reference Broussolle E, Dentresangle C, Landais P, Garcia-Larrea L, Pollak P, Croisile B, et al. The relation of putamen and caudate nucleus 18 F-Dopa uptake to motor and cognitive performances in Parkinson’s disease. J Neurol Sci. 1999;166:141–51.PubMedCrossRef Broussolle E, Dentresangle C, Landais P, Garcia-Larrea L, Pollak P, Croisile B, et al. The relation of putamen and caudate nucleus 18 F-Dopa uptake to motor and cognitive performances in Parkinson’s disease. J Neurol Sci. 1999;166:141–51.PubMedCrossRef
141.
go back to reference Brooks DJ, Ibanez V, Sawle GV, Quinn N, Lees AJ, Mathias CJ, et al. Differing patterns of striatal 18 F-dopa uptake in Parkinson’s disease, multiple system atrophy, and progressive supranuclear palsy. Ann Neurol. 1990;28:547–55.PubMedCrossRef Brooks DJ, Ibanez V, Sawle GV, Quinn N, Lees AJ, Mathias CJ, et al. Differing patterns of striatal 18 F-dopa uptake in Parkinson’s disease, multiple system atrophy, and progressive supranuclear palsy. Ann Neurol. 1990;28:547–55.PubMedCrossRef
142.
go back to reference Otsuka M, Ichiya Y, Hosokawa S, Kuwabara Y, Tahara T, Fukumura T, et al. Striatal blood flow, glucose metabolism and 18 F-dopa uptake: difference in Parkinson’s disease and atypical parkinsonism. J Neurol Neurosurg Psychiatry. 1991;54:898–904.PubMedPubMedCentralCrossRef Otsuka M, Ichiya Y, Hosokawa S, Kuwabara Y, Tahara T, Fukumura T, et al. Striatal blood flow, glucose metabolism and 18 F-dopa uptake: difference in Parkinson’s disease and atypical parkinsonism. J Neurol Neurosurg Psychiatry. 1991;54:898–904.PubMedPubMedCentralCrossRef
143.
go back to reference Ghaemi M, Hilker R, Rudolf J, Sobesky J, Heiss WD. Differentiating multiple system atrophy from parkinson’s disease: contribution of striatal and midbrain MRI volumetry and multi-tracer PET imaging. J Neurol Neurosurg Psychiatry. 2002;73:517–23.PubMedPubMedCentralCrossRef Ghaemi M, Hilker R, Rudolf J, Sobesky J, Heiss WD. Differentiating multiple system atrophy from parkinson’s disease: contribution of striatal and midbrain MRI volumetry and multi-tracer PET imaging. J Neurol Neurosurg Psychiatry. 2002;73:517–23.PubMedPubMedCentralCrossRef
144.
go back to reference Jin S, Oh M, Oh SJ, Oh JS, Lee SJ, Chung SJ, et al. Differential diagnosis of parkinsonism using dual-phase F-18 FP-CIT PET imaging. Nucl Med Mol Imaging (2010). 2013;47:44–51.CrossRef Jin S, Oh M, Oh SJ, Oh JS, Lee SJ, Chung SJ, et al. Differential diagnosis of parkinsonism using dual-phase F-18 FP-CIT PET imaging. Nucl Med Mol Imaging (2010). 2013;47:44–51.CrossRef
145.
go back to reference Fazio P, Svenningsson P, Forsberg A, Jönsson EG, Amini N, Nakao R, et al. Quantitative analysis of 18 F-(E)-N-(3-iodoprop-2-enyl)-2β-carbofluoroethoxy-3β-(4′-methyl-phenyl) nortropane binding to the dopamine transporter in Parkinson disease. J Nucl Med. 2015;56:714–20.PubMedCrossRef Fazio P, Svenningsson P, Forsberg A, Jönsson EG, Amini N, Nakao R, et al. Quantitative analysis of 18 F-(E)-N-(3-iodoprop-2-enyl)-2β-carbofluoroethoxy-3β-(4′-methyl-phenyl) nortropane binding to the dopamine transporter in Parkinson disease. J Nucl Med. 2015;56:714–20.PubMedCrossRef
146.
go back to reference Lee CS, Samii A, Sossi V, Ruth TJ, Schulzer M, Holden JE, et al. In vivo positron emission tomographic evidence for compensatory changes in presynaptic dopaminergic nerve terminals in Parkinson’s disease. Ann Neurol. 2000;47:493–503.PubMedCrossRef Lee CS, Samii A, Sossi V, Ruth TJ, Schulzer M, Holden JE, et al. In vivo positron emission tomographic evidence for compensatory changes in presynaptic dopaminergic nerve terminals in Parkinson’s disease. Ann Neurol. 2000;47:493–503.PubMedCrossRef
147.
go back to reference Okamura N, Villemagne VL, Drago J, Pejoska S, Dhamija RK, Mulligan RS, et al. In vivo measurement of vesicular monoamine transporter type 2 density in Parkinson disease with (18)F-AV-133. J Nucl Med. 2010;51:223–8.PubMedCrossRef Okamura N, Villemagne VL, Drago J, Pejoska S, Dhamija RK, Mulligan RS, et al. In vivo measurement of vesicular monoamine transporter type 2 density in Parkinson disease with (18)F-AV-133. J Nucl Med. 2010;51:223–8.PubMedCrossRef
148.
go back to reference Antonini A, Schwarz J, Oertel WH, Beer HF, Madeja UD, Leenders KL. [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. 1994;44:1325–9.PubMedCrossRef Antonini A, Schwarz J, Oertel WH, Beer HF, Madeja UD, Leenders KL. [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. 1994;44:1325–9.PubMedCrossRef
149.
go back to reference Rinne UK, Laihinen A, Rinne JO, Nagren 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.PubMedCrossRef Rinne UK, Laihinen A, Rinne JO, Nagren 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.PubMedCrossRef
150.
go back to reference Brooks DJ, Ibanez V, Sawle GV, Playford ED, Quinn N, Mathias CJ, 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.PubMedCrossRef Brooks DJ, Ibanez V, Sawle GV, Playford ED, Quinn N, Mathias CJ, 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.PubMedCrossRef
151.
go back to reference Antonini A, Leenders KL, Vontobel P, Maguire RP, Missimer J, Psylla M, et al. Complementary PET studies of striatal neuronal function in the differential diagnosis between multiple system atrophy and Parkinson’s disease. Brain. 1997;120:2187–95.PubMedCrossRef Antonini A, Leenders KL, Vontobel P, Maguire RP, Missimer J, Psylla M, et al. Complementary PET studies of striatal neuronal function in the differential diagnosis between multiple system atrophy and Parkinson’s disease. Brain. 1997;120:2187–95.PubMedCrossRef
152.
go back to reference Van Laere K, Clerinx K, D’Hondt E, de Groot T, Vandenberghe W. Combined Striatal binding and cerebral influx analysis of dynamic 11C-raclopride PET improves early differentiation between multiple-system atrophy and Parkinson disease. J Nucl Med. 2010;51:588–95.PubMedCrossRef Van Laere K, Clerinx K, D’Hondt E, de Groot T, Vandenberghe W. Combined Striatal binding and cerebral influx analysis of dynamic 11C-raclopride PET improves early differentiation between multiple-system atrophy and Parkinson disease. J Nucl Med. 2010;51:588–95.PubMedCrossRef
153.
go back to reference Zhao P, Zhang B, Gao S. 18F-FDG PET study on the idiopathic Parkinson’s disease from several parkinsonian-plus syndromes. Parkinsonism Relat Disord. 2012;18 Suppl 1:S60–2.PubMedCrossRef Zhao P, Zhang B, Gao S. 18F-FDG PET study on the idiopathic Parkinson’s disease from several parkinsonian-plus syndromes. Parkinsonism Relat Disord. 2012;18 Suppl 1:S60–2.PubMedCrossRef
154.
go back to reference Akdemir UO, Tokcaer AB, Karakus A, Kapucu LO. Brain 18 F-FDG PET imaging in the differential diagnosis of parkinsonism. Clin Nucl Med. 2014;93:e220–6.CrossRef Akdemir UO, Tokcaer AB, Karakus A, Kapucu LO. Brain 18 F-FDG PET imaging in the differential diagnosis of parkinsonism. Clin Nucl Med. 2014;93:e220–6.CrossRef
156.
go back to reference Juh R, Kim J, Moon D, Choe B, Suh T. Different metabolic patterns analysis of Parkinsonism on the 18 F-FDG PET. Eur J Radiol. 2004;51:223–33.PubMedCrossRef Juh R, Kim J, Moon D, Choe B, Suh T. Different metabolic patterns analysis of Parkinsonism on the 18 F-FDG PET. Eur J Radiol. 2004;51:223–33.PubMedCrossRef
157.
go back to reference Eckert T, Barnes A, Dhawan V, Frucht S, Gordon MF, Feigin AS, et al. FDG PET in the differential diagnosis of parkinsonian disorders. Neuroimage. 2005;26:912–21.PubMedCrossRef Eckert T, Barnes A, Dhawan V, Frucht S, Gordon MF, Feigin AS, et al. FDG PET in the differential diagnosis of parkinsonian disorders. Neuroimage. 2005;26:912–21.PubMedCrossRef
158.
go back to reference Baudrexel S, Seifried C, Penndorf B, Klein JC, Middendorp M, Steinmetz H, et al. The value of putaminal diffusion imaging versus 18-fluorodeoxyglucose positron emission tomography for the differential diagnosis of the Parkinson variant of multiple system atrophy. Mov Disord. 2014;29:380–7.PubMedCrossRef Baudrexel S, Seifried C, Penndorf B, Klein JC, Middendorp M, Steinmetz H, et al. The value of putaminal diffusion imaging versus 18-fluorodeoxyglucose positron emission tomography for the differential diagnosis of the Parkinson variant of multiple system atrophy. Mov Disord. 2014;29:380–7.PubMedCrossRef
159.
go back to reference Hosaka K, Ishii K, Sakamoto S, Mori T, Sasaki M, Hirono N, et al. Voxel-based comparison of regional cerebral glucose metabolism between PSP and corticobasal degeneration. J Neurol Sci. 2002;199:67–71.PubMedCrossRef Hosaka K, Ishii K, Sakamoto S, Mori T, Sasaki M, Hirono N, et al. Voxel-based comparison of regional cerebral glucose metabolism between PSP and corticobasal degeneration. J Neurol Sci. 2002;199:67–71.PubMedCrossRef
160.
go back to reference Botha H, Whitwell JL, Madhaven A, Senjem ML, Lowe V, Josephs KA. The pimple sign of progressive supranuclear palsy syndrome. Park Relat Disord. 2014;20:180–5.CrossRef Botha H, Whitwell JL, Madhaven A, Senjem ML, Lowe V, Josephs KA. The pimple sign of progressive supranuclear palsy syndrome. Park Relat Disord. 2014;20:180–5.CrossRef
161.
go back to reference Niethammer M, Tang CC, Feigin A, Allen PJ, Heinen L, Hellwig S, et al. A disease-specific metabolic brain network associated with corticobasal degeneration. Brain. 2014;137:3036–46.PubMedPubMedCentralCrossRef Niethammer M, Tang CC, Feigin A, Allen PJ, Heinen L, Hellwig S, et al. A disease-specific metabolic brain network associated with corticobasal degeneration. Brain. 2014;137:3036–46.PubMedPubMedCentralCrossRef
162.
go back to reference Tripathi M, Dhawan V, Peng S, Kushwaha S, Batla A, Jaimini A, et al. Differential diagnosis of parkinsonian syndromes using F-18 fluorodeoxyglucose positron emission tomography. Neuroradiology. 2013;55:483–92.PubMedCrossRef Tripathi M, Dhawan V, Peng S, Kushwaha S, Batla A, Jaimini A, et al. Differential diagnosis of parkinsonian syndromes using F-18 fluorodeoxyglucose positron emission tomography. Neuroradiology. 2013;55:483–92.PubMedCrossRef
163.
go back to reference Mosconi L, Tsui WH, Herholz K, Pupi A, Drzezga A, Lucignani G, et al. Multicenter standardized 18 F-FDG PET diagnosis of mild cognitive impairment, Alzheimer’s disease, and other dementias. J Nucl Med. 2008;49:390–8.PubMedPubMedCentralCrossRef Mosconi L, Tsui WH, Herholz K, Pupi A, Drzezga A, Lucignani G, et al. Multicenter standardized 18 F-FDG PET diagnosis of mild cognitive impairment, Alzheimer’s disease, and other dementias. J Nucl Med. 2008;49:390–8.PubMedPubMedCentralCrossRef
164.
go back to reference Lim SM, Katsifis A, Villemagne VL, Best R, Jones G, Saling M, et al. The 18 F-FDG PET cingulate island sign and comparison to 123I-beta-CIT SPECT for diagnosis of dementia with Lewy bodies. J Nucl Med. 2009;50:1638–45.PubMedCrossRef Lim SM, Katsifis A, Villemagne VL, Best R, Jones G, Saling M, et al. The 18 F-FDG PET cingulate island sign and comparison to 123I-beta-CIT SPECT for diagnosis of dementia with Lewy bodies. J Nucl Med. 2009;50:1638–45.PubMedCrossRef
165.
go back to reference Yong SW, Yoon JK, An YS, Lee PH. A comparison of cerebral glucose metabolism in Parkinson’s disease, Parkinson’s disease dementia and dementia with Lewy bodies. Eur J Neurol. 2007;14:1357–62.PubMedCrossRef Yong SW, Yoon JK, An YS, Lee PH. A comparison of cerebral glucose metabolism in Parkinson’s disease, Parkinson’s disease dementia and dementia with Lewy bodies. Eur J Neurol. 2007;14:1357–62.PubMedCrossRef
167.
go back to reference Eckert T, Van Laere K, Tang C, Lewis DE, Edwards C, Santens P, et al. Quantification of Parkinson’s disease-related network expression with ECD SPECT. Eur J Nucl Med Mol Imaging. 2007;34:496–501.PubMedCrossRef Eckert T, Van Laere K, Tang C, Lewis DE, Edwards C, Santens P, et al. Quantification of Parkinson’s disease-related network expression with ECD SPECT. Eur J Nucl Med Mol Imaging. 2007;34:496–501.PubMedCrossRef
168.
go back to reference Huang C, Mattis P, Tang C, Perrine K, Carbon M, Eidelberg D. Metabolic brain networks associated with cognitive function in Parkinson’s disease. Neuroimage. 2007;34:714–23.PubMedCrossRef Huang C, Mattis P, Tang C, Perrine K, Carbon M, Eidelberg D. Metabolic brain networks associated with cognitive function in Parkinson’s disease. Neuroimage. 2007;34:714–23.PubMedCrossRef
169.
go back to reference Eckert T, Tang C, Ma Y, Brown N, Lin T, Frucht S, et al. Abnormal metabolic networks in atypical parkinsonism. Mov Disord. 2008;23:727–33.PubMedCrossRef Eckert T, Tang C, Ma Y, Brown N, Lin T, Frucht S, et al. Abnormal metabolic networks in atypical parkinsonism. Mov Disord. 2008;23:727–33.PubMedCrossRef
170.
go back to reference Edison P, Rowe CC, Rinne JO, Ng S, Ahmed I, Kemppainen N, et al. Amyloid load in Parkinson’s disease dementia and Lewy body dementia measured with [11C]PIB positron emission tomography. J Neurol Neurosurg Psychiatry. 2008;79:1331–8.PubMedCrossRef Edison P, Rowe CC, Rinne JO, Ng S, Ahmed I, Kemppainen N, et al. Amyloid load in Parkinson’s disease dementia and Lewy body dementia measured with [11C]PIB positron emission tomography. J Neurol Neurosurg Psychiatry. 2008;79:1331–8.PubMedCrossRef
171.
go back to reference Gomperts SN, Locascio JJ, Marquie M, Santarlasci AL, Rentz DM, Maye J, et al. Brain amyloid and cognition in Lewy body diseases. Mov Disord. 2012;27:965–73.PubMedPubMedCentralCrossRef Gomperts SN, Locascio JJ, Marquie M, Santarlasci AL, Rentz DM, Maye J, et al. Brain amyloid and cognition in Lewy body diseases. Mov Disord. 2012;27:965–73.PubMedPubMedCentralCrossRef
172.
go back to reference Foster ER, Campbell MC, Burack MA, Hartlein J, Flores HP, Cairns NJ, et al. Amyloid imaging of Lewy body-associated disorders. Mov Disord. 2010;25:2516–23.PubMedPubMedCentralCrossRef Foster ER, Campbell MC, Burack MA, Hartlein J, Flores HP, Cairns NJ, et al. Amyloid imaging of Lewy body-associated disorders. Mov Disord. 2010;25:2516–23.PubMedPubMedCentralCrossRef
173.
go back to reference Donaghy P, Thomas AJ, O’Brien JT. Amyloid PET imaging in lewy body disorders. Am J Geriatr Psychiatry. 2015;23:23–37.PubMedCrossRef Donaghy P, Thomas AJ, O’Brien JT. Amyloid PET imaging in lewy body disorders. Am J Geriatr Psychiatry. 2015;23:23–37.PubMedCrossRef
174.
go back to reference Harding AJ, Halliday GM. Cortical Lewy body pathology in the diagnosis of dementia. Acta Neuropathol. 2001;102:355–63.PubMed Harding AJ, Halliday GM. Cortical Lewy body pathology in the diagnosis of dementia. Acta Neuropathol. 2001;102:355–63.PubMed
175.
go back to reference Irwin DJ, White MT, Toledo JB, Xie SX, Robinson JL, Van Deerlin V, et al. Neuropathologic substrates of Parkinson disease dementia. Ann Neurol. 2012;72:587–98.PubMedPubMedCentralCrossRef Irwin DJ, White MT, Toledo JB, Xie SX, Robinson JL, Van Deerlin V, et al. Neuropathologic substrates of Parkinson disease dementia. Ann Neurol. 2012;72:587–98.PubMedPubMedCentralCrossRef
176.
go back to reference Claassen DO, Lowe VJ, Peller PJ, Petersen RC, Josephs KA. Amyloid and glucose imaging in dementia with Lewy bodies and multiple systems atrophy. Park Relat Disord. 2011;17:160–5.CrossRef Claassen DO, Lowe VJ, Peller PJ, Petersen RC, Josephs KA. Amyloid and glucose imaging in dementia with Lewy bodies and multiple systems atrophy. Park Relat Disord. 2011;17:160–5.CrossRef
177.
go back to reference Villemagne VL, Ong K, Mulligan RS, Holl G, Pejoska S, Jones G, et al. Amyloid imaging with (18)F-florbetaben in Alzheimer disease and other dementias. J Nucl Med. 2011;52:1210–7.PubMedCrossRef Villemagne VL, Ong K, Mulligan RS, Holl G, Pejoska S, Jones G, et al. Amyloid imaging with (18)F-florbetaben in Alzheimer disease and other dementias. J Nucl Med. 2011;52:1210–7.PubMedCrossRef
178.
go back to reference Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe CC. Tau imaging: early progress and future directions. Lancet Neurol. 2015;14:114–24.PubMedCrossRef Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe CC. Tau imaging: early progress and future directions. Lancet Neurol. 2015;14:114–24.PubMedCrossRef
179.
go back to reference Kepe V, Bordelon Y, Boxer A, Huang SC, Liu J, Thiede FC, et al. PET imaging of neuropathology in tauopathies: progressive Supranuclear palsy. J Alzheimer’s Dis. 2013;36:145–53. Kepe V, Bordelon Y, Boxer A, Huang SC, Liu J, Thiede FC, et al. PET imaging of neuropathology in tauopathies: progressive Supranuclear palsy. J Alzheimer’s Dis. 2013;36:145–53.
180.
go back to reference Maruyama M, Shimada H, Suhara T, Shinotoh H, Ji B, Maeda J, et al. Imaging of tau pathology in a tauopathy mouse model and in Alzheimer patients compared to normal controls. Neuron. 2013;79:1094–108.PubMedCrossRef Maruyama M, Shimada H, Suhara T, Shinotoh H, Ji B, Maeda J, et al. Imaging of tau pathology in a tauopathy mouse model and in Alzheimer patients compared to normal controls. Neuron. 2013;79:1094–108.PubMedCrossRef
181.
go back to reference Xia C-F, Arteaga J, Chen G, Gangadharmath U, Gomez LF, Kasi D, et al. [18 F]T807, a novel tau positron emission tomography imaging agent for Alzheimer’s disease. Alzheimer’s Dement. 2013;9:666–76.CrossRef Xia C-F, Arteaga J, Chen G, Gangadharmath U, Gomez LF, Kasi D, et al. [18 F]T807, a novel tau positron emission tomography imaging agent for Alzheimer’s disease. Alzheimer’s Dement. 2013;9:666–76.CrossRef
182.
go back to reference Marquié M, Normandin MD, Vanderburg CR, Costantino IM, Bien EA, Rycyna LG, et al. Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol. 2015;78:787–800.PubMedPubMedCentralCrossRef Marquié M, Normandin MD, Vanderburg CR, Costantino IM, Bien EA, Rycyna LG, et al. Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol. 2015;78:787–800.PubMedPubMedCentralCrossRef
183.
go back to reference Hansen AK, Knudsen K, Lillethorup TP, Landau AM, Parbo P, Fedorova T, et al. In vivo imaging of neuromelanin in Parkinson’s disease using 18 F-AV-1451 PET. Brain. 2016;139:2039–49.PubMedCrossRef Hansen AK, Knudsen K, Lillethorup TP, Landau AM, Parbo P, Fedorova T, et al. In vivo imaging of neuromelanin in Parkinson’s disease using 18 F-AV-1451 PET. Brain. 2016;139:2039–49.PubMedCrossRef
184.
go back to reference Cho H, Choi JY, Hwang MS, Lee SH, Ryu YH, Lee MS, et al. Subcortical (18) F-AV-1451 binding patterns in progressive supranuclear palsy. Mov Disord. 2016. doi:10.1002/mds.26844 [Epub ahead of print]. Cho H, Choi JY, Hwang MS, Lee SH, Ryu YH, Lee MS, et al. Subcortical (18) F-AV-1451 binding patterns in progressive supranuclear palsy. Mov Disord. 2016. doi:10.​1002/​mds.​26844 [Epub ahead of print].
185.
go back to reference Smith R, Schain M, Nilsson C, Strandberg O, Olsson T, Hägerström D, et al. Increased basal ganglia binding of (18) F-AV-1451 in patients with progressive supranuclear palsy. Mov Disord. 2016. doi:10.1002/mds.26813 [Epub ahead of print].PubMedCentral Smith R, Schain M, Nilsson C, Strandberg O, Olsson T, Hägerström D, et al. Increased basal ganglia binding of (18) F-AV-1451 in patients with progressive supranuclear palsy. Mov Disord. 2016. doi:10.​1002/​mds.​26813 [Epub ahead of print].PubMedCentral
186.
go back to reference Whitwell JL, Lowe VJ, Tosakulwong N, Weigand SD, Senjem ML, Schwarz CG, et al. [(18) F] AV-1451 tau positron emission tomography in progressive supranuclear palsy. Mov Disord. 2016. doi:10.1002/mds.26834 [Epub ahead of print].PubMed Whitwell JL, Lowe VJ, Tosakulwong N, Weigand SD, Senjem ML, Schwarz CG, et al. [(18) F] AV-1451 tau positron emission tomography in progressive supranuclear palsy. Mov Disord. 2016. doi:10.​1002/​mds.​26834 [Epub ahead of print].PubMed
187.
go back to reference Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009;8:382–97.PubMedCrossRef Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009;8:382–97.PubMedCrossRef
188.
go back to reference Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol Dis. 2006;21:404–12.PubMedCrossRef Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol Dis. 2006;21:404–12.PubMedCrossRef
189.
go back to reference Ouchi Y, Yoshikawa E, Sekine Y, Futatsubashi M, Kanno T, Ogusu T, et al. Microglial activation and dopamine terminal loss in early Parkinson’s disease. Ann Neurol. 2005;57:168–75.PubMedCrossRef Ouchi Y, Yoshikawa E, Sekine Y, Futatsubashi M, Kanno T, Ogusu T, et al. Microglial activation and dopamine terminal loss in early Parkinson’s disease. Ann Neurol. 2005;57:168–75.PubMedCrossRef
190.
go back to reference Edison P, Ahmed I, Fan Z, Hinz R, Gelosa G, Ray Chaudhuri K, et al. Microglia, amyloid, and glucose metabolism in Parkinson’s disease with and without dementia. Neuropsychopharmacology. 2013;38:938–49.PubMedPubMedCentralCrossRef Edison P, Ahmed I, Fan Z, Hinz R, Gelosa G, Ray Chaudhuri K, et al. Microglia, amyloid, and glucose metabolism in Parkinson’s disease with and without dementia. Neuropsychopharmacology. 2013;38:938–49.PubMedPubMedCentralCrossRef
191.
go back to reference Gerhard A, Banati RB, Goerres GB, Cagnin A, Myers R, Gunn RN, et al. [11C](R)-PK11195 PET imaging of microglial activation in multiple system atrophy. Neurology. 2003;61:686–9.PubMedCrossRef Gerhard A, Banati RB, Goerres GB, Cagnin A, Myers R, Gunn RN, et al. [11C](R)-PK11195 PET imaging of microglial activation in multiple system atrophy. Neurology. 2003;61:686–9.PubMedCrossRef
192.
go back to reference Gerhard A, Watts J, Trender-Gerhard I, Turkheimer F, Banati RB, Bhatia K, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in corticobasal degeneration. Mov Disord. 2004;19:1221–6.PubMedCrossRef Gerhard A, Watts J, Trender-Gerhard I, Turkheimer F, Banati RB, Bhatia K, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in corticobasal degeneration. Mov Disord. 2004;19:1221–6.PubMedCrossRef
193.
go back to reference Fan Z, Aman Y, Ahmed I, Chetelat G, Landeau B, Ray Chaudhuri K, et al. Influence of microglial activation on neuronal function in Alzheimer’s and Parkinson’s disease dementia. Alzheimer’s Dement J Alzheimer’s Assoc. 2015;11:608–21. e7.CrossRef Fan Z, Aman Y, Ahmed I, Chetelat G, Landeau B, Ray Chaudhuri K, et al. Influence of microglial activation on neuronal function in Alzheimer’s and Parkinson’s disease dementia. Alzheimer’s Dement J Alzheimer’s Assoc. 2015;11:608–21. e7.CrossRef
194.
go back to reference Yokokura M, Terada T, Bunai T, Nakaizumi K, Takebayashi K, Iwata Y, et al. Depiction of microglial activation in aging and dementia: Positron emission tomography with [11C]DPA713 versus [11C](R)PK11195. J Cereb Blood Flow Metab. 2016. [Epub ahead of print]. Yokokura M, Terada T, Bunai T, Nakaizumi K, Takebayashi K, Iwata Y, et al. Depiction of microglial activation in aging and dementia: Positron emission tomography with [11C]DPA713 versus [11C](R)PK11195. J Cereb Blood Flow Metab. 2016. [Epub ahead of print].
195.
go back to reference Gasnier B, Roisin MP, Scherman D, Coornaert S, Desplanches G, Henry JP. Uptake of meta-iodobenzylguanidine by bovine chromaffin granule membranes. Mol Pharmacol. 1986;29:275–80.PubMed Gasnier B, Roisin MP, Scherman D, Coornaert S, Desplanches G, Henry JP. Uptake of meta-iodobenzylguanidine by bovine chromaffin granule membranes. Mol Pharmacol. 1986;29:275–80.PubMed
196.
go back to reference Orimo S, Yogo M, Nakamura T, Suzuki M, Watanabe H. 123I-meta-iodobenzylguanidine (MIBG) cardiac scintigraphy in α-synucleinopathies. Ageing Res Rev. 2016;30:122–33.PubMedCrossRef Orimo S, Yogo M, Nakamura T, Suzuki M, Watanabe H. 123I-meta-iodobenzylguanidine (MIBG) cardiac scintigraphy in α-synucleinopathies. Ageing Res Rev. 2016;30:122–33.PubMedCrossRef
197.
go back to reference Yoshita M, Taki J, Yamada M. A clinical role for [(123) I] MIBG myocardial scintigraphy in the distinction between dementia of the Alzheimer’s-type and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2001;71:583–8.PubMedPubMedCentralCrossRef Yoshita M, Taki J, Yamada M. A clinical role for [(123) I] MIBG myocardial scintigraphy in the distinction between dementia of the Alzheimer’s-type and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2001;71:583–8.PubMedPubMedCentralCrossRef
198.
go back to reference Braune S, Reinhardt M, Schnitzer R, Riedel A, Lücking CH. Cardiac uptake of [123I]MIBG separates Parkinson’s disease from multiple system atrophy. Neurology. 1999;53:1020–5.PubMedCrossRef Braune S, Reinhardt M, Schnitzer R, Riedel A, Lücking CH. Cardiac uptake of [123I]MIBG separates Parkinson’s disease from multiple system atrophy. Neurology. 1999;53:1020–5.PubMedCrossRef
199.
go back to reference Yoshita M. Differentiation of idiopathic Parkinson’s disease from striatonigral degeneration and progressive supranuclear palsy using iodine-123 meta-iodobenzylguanidine myocardial scintigraphy. J Neurol Sci. 1998;155:60–7.PubMedCrossRef Yoshita M. Differentiation of idiopathic Parkinson’s disease from striatonigral degeneration and progressive supranuclear palsy using iodine-123 meta-iodobenzylguanidine myocardial scintigraphy. J Neurol Sci. 1998;155:60–7.PubMedCrossRef
200.
go back to reference Raffel DM, Koeppe RA, Little R, Wang C-N, Liu S, Junck L, et al. PET measurement of cardiac and nigrostriatal denervation in Parkinsonian syndromes. J Nucl Med. 2006;47:1769–77.PubMed Raffel DM, Koeppe RA, Little R, Wang C-N, Liu S, Junck L, et al. PET measurement of cardiac and nigrostriatal denervation in Parkinsonian syndromes. J Nucl Med. 2006;47:1769–77.PubMed
201.
go back to reference Oka H, Toyoda C, Yogo M, Mochio S. Cardiovascular dysautonomia in de novo Parkinson’s disease without orthostatic hypotension. Eur J Neurol. 2011;18:286–92.PubMedCrossRef Oka H, Toyoda C, Yogo M, Mochio S. Cardiovascular dysautonomia in de novo Parkinson’s disease without orthostatic hypotension. Eur J Neurol. 2011;18:286–92.PubMedCrossRef
202.
go back to reference Taki J, Nakajima K, Hwang EH, Matsunari I, Komai K, Yoshita M, et al. Peripheral sympathetic dysfunction in patients with Parkinson’s disease without autonomic failure is heart selective and disease specific. EurJ Nucl Med. 2000;27:566–73.CrossRef Taki J, Nakajima K, Hwang EH, Matsunari I, Komai K, Yoshita M, et al. Peripheral sympathetic dysfunction in patients with Parkinson’s disease without autonomic failure is heart selective and disease specific. EurJ Nucl Med. 2000;27:566–73.CrossRef
203.
go back to reference Takatsu H, Nishida H, Matsuo H, Watanabe S, Nagashima K, Wada H, et al. Cardiac sympathetic denervation from the early stage of Parkinson’s disease: clinical and experimental studies with radiolabeled MIBG. J Nucl Med. 2000;41:71–7.PubMed Takatsu H, Nishida H, Matsuo H, Watanabe S, Nagashima K, Wada H, et al. Cardiac sympathetic denervation from the early stage of Parkinson’s disease: clinical and experimental studies with radiolabeled MIBG. J Nucl Med. 2000;41:71–7.PubMed
204.
go back to reference Taki J, Yoshita M, Yamada M, Tonami N. Significance of 123I-MIBG scintigraphy as a pathophysiological indicator in the assessment of Parkinson’s disease and related disorders: it can be a specific marker for Lewy body disease. Ann Nucl Med. 2004;18:453–61.PubMedCrossRef Taki J, Yoshita M, Yamada M, Tonami N. Significance of 123I-MIBG scintigraphy as a pathophysiological indicator in the assessment of Parkinson’s disease and related disorders: it can be a specific marker for Lewy body disease. Ann Nucl Med. 2004;18:453–61.PubMedCrossRef
205.
go back to reference Hanyu H, Shimizu S, Hirao K, Sakurai H, Iwamoto T, Chikamori T, et al. The role of 123I-metaiodobenzylguanidine myocardial scintigraphy in the diagnosis of Lewy body disease in patients with dementia in a memory clinic. Dement Geriatr Cogn Disord. 2006;22:379–84.PubMedCrossRef Hanyu H, Shimizu S, Hirao K, Sakurai H, Iwamoto T, Chikamori T, et al. The role of 123I-metaiodobenzylguanidine myocardial scintigraphy in the diagnosis of Lewy body disease in patients with dementia in a memory clinic. Dement Geriatr Cogn Disord. 2006;22:379–84.PubMedCrossRef
206.
go back to reference Hanyu H, Shimizu S, Hirao K, Kanetaka H, Iwamoto T, Chikamori T, et al. Comparative value of brain perfusion SPECT and [123I] MIBG myocardial scintigraphy in distinguishing between dementia with Lewy bodies and Alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2006;33:248–53.PubMedCrossRef Hanyu H, Shimizu S, Hirao K, Kanetaka H, Iwamoto T, Chikamori T, et al. Comparative value of brain perfusion SPECT and [123I] MIBG myocardial scintigraphy in distinguishing between dementia with Lewy bodies and Alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2006;33:248–53.PubMedCrossRef
207.
go back to reference Wada-Isoe K, Kitayama M, Nakaso K, Nakashima K. Diagnostic markers for diagnosing dementia with Lewy bodies: CSF and MIBG cardiac scintigraphy study. J Neurol Sci. 2007;260:33–7.PubMedCrossRef Wada-Isoe K, Kitayama M, Nakaso K, Nakashima K. Diagnostic markers for diagnosing dementia with Lewy bodies: CSF and MIBG cardiac scintigraphy study. J Neurol Sci. 2007;260:33–7.PubMedCrossRef
208.
go back to reference King AE, Mintz J, Royall DR. Meta-analysis of 123I-MIBG cardiac scintigraphy for the diagnosis of Lewy body-related disorders. Mov Disord. 2011;26:1218–24.PubMedCrossRef King AE, Mintz J, Royall DR. Meta-analysis of 123I-MIBG cardiac scintigraphy for the diagnosis of Lewy body-related disorders. Mov Disord. 2011;26:1218–24.PubMedCrossRef
209.
go back to reference Treglia G, Cason E, Stefanelli A, Cocciolillo F, Di Giuda D, Fagioli G, et al. MIBG scintigraphy in differential diagnosis of parkinsonism: a meta-analysis. Clin Auton Res. 2012;22:43–55.PubMedCrossRef Treglia G, Cason E, Stefanelli A, Cocciolillo F, Di Giuda D, Fagioli G, et al. MIBG scintigraphy in differential diagnosis of parkinsonism: a meta-analysis. Clin Auton Res. 2012;22:43–55.PubMedCrossRef
210.
go back to reference Südmeyer M, Antke C, Zizek T, Beu M, Nikolaus S, Wojtecki L, et al. Diagnostic accuracy of combined FP-CIT, IBZM, and MIBG scintigraphy in the differential diagnosis of degenerative parkinsonism: a multidimensional statistical approach. J Nucl Med. 2011;52:733–40.PubMedCrossRef Südmeyer M, Antke C, Zizek T, Beu M, Nikolaus S, Wojtecki L, et al. Diagnostic accuracy of combined FP-CIT, IBZM, and MIBG scintigraphy in the differential diagnosis of degenerative parkinsonism: a multidimensional statistical approach. J Nucl Med. 2011;52:733–40.PubMedCrossRef
211.
go back to reference Sengoku R, Matsushima S, Bono K, Sakuta K, Yamazaki M, Miyagawa S, et al. Olfactory function combined with morphology distinguishes Parkinson’s disease. Parkinsonism Relat Disord. 2015;21:771–7.PubMedCrossRef Sengoku R, Matsushima S, Bono K, Sakuta K, Yamazaki M, Miyagawa S, et al. Olfactory function combined with morphology distinguishes Parkinson’s disease. Parkinsonism Relat Disord. 2015;21:771–7.PubMedCrossRef
212.
go back to reference Sakata K, Iida K, Mochizuki N, Ito M, Nakaya Y. Physiological changes in human cardiac sympathetic innervation and activity assessed by (123) I-metaiodobenzylguanidine (MIGB) imaging. Circ J. 2009;73:310–5.PubMedCrossRef Sakata K, Iida K, Mochizuki N, Ito M, Nakaya Y. Physiological changes in human cardiac sympathetic innervation and activity assessed by (123) I-metaiodobenzylguanidine (MIGB) imaging. Circ J. 2009;73:310–5.PubMedCrossRef
213.
go back to reference Berg D, Siefker C, Becker G. Echogenicity of the substantia nigra in Parkinson’s disease and its relation to clinical findings. J Neurol. 2001;248:684–9.PubMedCrossRef Berg D, Siefker C, Becker G. Echogenicity of the substantia nigra in Parkinson’s disease and its relation to clinical findings. J Neurol. 2001;248:684–9.PubMedCrossRef
214.
go back to reference Gaenslen A, Unmuth B, Godau J, Liepelt I, Di Santo A, Schweitzer KJ, et al. The specificity and sensitivity of transcranial ultrasound in the differential diagnosis of Parkinson’s disease: a prospective blinded study. Lancet Neurol. 2008;7:417–24.PubMedCrossRef Gaenslen A, Unmuth B, Godau J, Liepelt I, Di Santo A, Schweitzer KJ, et al. The specificity and sensitivity of transcranial ultrasound in the differential diagnosis of Parkinson’s disease: a prospective blinded study. Lancet Neurol. 2008;7:417–24.PubMedCrossRef
215.
go back to reference Behnke S, Berg D, Naumann M, Becker G. Differentiation of Parkinson’s disease and atypical parkinsonian syndromes by transcranial ultrasound. J Neurol Neurosurg Psychiatry. 2005;76:423–5.PubMedPubMedCentralCrossRef Behnke S, Berg D, Naumann M, Becker G. Differentiation of Parkinson’s disease and atypical parkinsonian syndromes by transcranial ultrasound. J Neurol Neurosurg Psychiatry. 2005;76:423–5.PubMedPubMedCentralCrossRef
216.
go back to reference Becker G, Seufert J, Bogdahn U, Reichmann H, Reiners K. Degeneration of substantia nigra in chronic Parkinson’s disease visualized by transcranial color-coded real-time sonography. Neurology. 1995;45:182–4.PubMedCrossRef Becker G, Seufert J, Bogdahn U, Reichmann H, Reiners K. Degeneration of substantia nigra in chronic Parkinson’s disease visualized by transcranial color-coded real-time sonography. Neurology. 1995;45:182–4.PubMedCrossRef
217.
go back to reference Berg D, Merz B, Reiners K, Naumann M, Becker G. Five-year follow-up study of hyperechogenicity of the substantia nigra in Parkinson’s disease. Mov Disord. 2005;20:383–5.PubMedCrossRef Berg D, Merz B, Reiners K, Naumann M, Becker G. Five-year follow-up study of hyperechogenicity of the substantia nigra in Parkinson’s disease. Mov Disord. 2005;20:383–5.PubMedCrossRef
218.
go back to reference Spiegel J, Hellwig D, Möllers MO, Behnke S, Jost W, Fassbender K, et al. Transcranial sonography and [123I]FP-CIT SPECT disclose complementary aspects of Parkinson’s disease. Brain. 2006;129:1188–93.PubMedCrossRef Spiegel J, Hellwig D, Möllers MO, Behnke S, Jost W, Fassbender K, et al. Transcranial sonography and [123I]FP-CIT SPECT disclose complementary aspects of Parkinson’s disease. Brain. 2006;129:1188–93.PubMedCrossRef
219.
go back to reference Berg D, Becker G, Zeiler B, Tucha O, Hofmann E, Preier M, et al. Vulnerability of the nigrostriatal system as detected by transcranial ultrasound. Neurology. 1999;53:1026–31.PubMedCrossRef Berg D, Becker G, Zeiler B, Tucha O, Hofmann E, Preier M, et al. Vulnerability of the nigrostriatal system as detected by transcranial ultrasound. Neurology. 1999;53:1026–31.PubMedCrossRef
220.
go back to reference Berg D, Roggendorf W, Schröder U, Klein R, Tatschner T, Benz P, et al. Echogenicity of the substantia nigra: association with increased iron content and marker for susceptibility to nigrostriatal injury. Arch Neurol. 2002;59:999–1005.PubMedCrossRef Berg D, Roggendorf W, Schröder U, Klein R, Tatschner T, Benz P, et al. Echogenicity of the substantia nigra: association with increased iron content and marker for susceptibility to nigrostriatal injury. Arch Neurol. 2002;59:999–1005.PubMedCrossRef
221.
go back to reference Walter U, Niehaus L, Probst T, Benecke R, Meyer BU, Dressler D. Brain parenchyma sonography discriminates Parkinson’s disease and atypical parkinsonian syndromes. Neurology. 2003;60:74–7.PubMedCrossRef Walter U, Niehaus L, Probst T, Benecke R, Meyer BU, Dressler D. Brain parenchyma sonography discriminates Parkinson’s disease and atypical parkinsonian syndromes. Neurology. 2003;60:74–7.PubMedCrossRef
222.
go back to reference Stockner H, Sojer M, Seppi KK, Mueller J, Wenning GK, Schmidauer C, et al. Midbrain sonography in patients with essential tremor. Mov Disord. 2007;22:414–7.PubMedCrossRef Stockner H, Sojer M, Seppi KK, Mueller J, Wenning GK, Schmidauer C, et al. Midbrain sonography in patients with essential tremor. Mov Disord. 2007;22:414–7.PubMedCrossRef
223.
go back to reference Walter U, Dressler D, Wolters A, Wittstock M, Greim B, Benecke R. Sonographic discrimination of dementia with Lewy bodies and Parkinson’s disease with dementia. J Neurol. 2006;253:448–54.PubMedCrossRef Walter U, Dressler D, Wolters A, Wittstock M, Greim B, Benecke R. Sonographic discrimination of dementia with Lewy bodies and Parkinson’s disease with dementia. J Neurol. 2006;253:448–54.PubMedCrossRef
224.
go back to reference Walter U, Dressler D, Wolters A, Probst T, Grossmann A, Benecke R. Sonographic discrimination of corticobasal degeneration vs progressive supranuclear palsy. Neurology. 2004;63:504–9.PubMedCrossRef Walter U, Dressler D, Wolters A, Probst T, Grossmann A, Benecke R. Sonographic discrimination of corticobasal degeneration vs progressive supranuclear palsy. Neurology. 2004;63:504–9.PubMedCrossRef
225.
go back to reference Sadowski K, Serafin-Król M, Szlachta K, Friedman A. Basal ganglia echogenicity in tauopathies. J Neural Transm (Vienna). 2015;122:863–5.CrossRef Sadowski K, Serafin-Król M, Szlachta K, Friedman A. Basal ganglia echogenicity in tauopathies. J Neural Transm (Vienna). 2015;122:863–5.CrossRef
226.
go back to reference Walter U, Dressler D, Probst T, Wolters A, Abu-Mugheisib M, Wittstock M, et al. Transcranial brain sonography findings in discriminating between parkinsonism and idiopathic Parkinson disease. Arch Neurol. 2007;64:1635–40.PubMedCrossRef Walter U, Dressler D, Probst T, Wolters A, Abu-Mugheisib M, Wittstock M, et al. Transcranial brain sonography findings in discriminating between parkinsonism and idiopathic Parkinson disease. Arch Neurol. 2007;64:1635–40.PubMedCrossRef
227.
go back to reference Ebentheuer J, Canelo M, Trautmann E, Trenkwalder C. Substantia nigra echogenicity in progressive supranuclear palsy. Mov Disord. 2010;25:773–7.PubMedCrossRef Ebentheuer J, Canelo M, Trautmann E, Trenkwalder C. Substantia nigra echogenicity in progressive supranuclear palsy. Mov Disord. 2010;25:773–7.PubMedCrossRef
Metadata
Title
Imaging biomarkers in Parkinson’s disease and Parkinsonian syndromes: current and emerging concepts
Authors
Usman Saeed
Jordana Compagnone
Richard I. Aviv
Antonio P. Strafella
Sandra E. Black
Anthony E. Lang
Mario Masellis
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Translational Neurodegeneration / Issue 1/2017
Electronic ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-017-0076-6

Other articles of this Issue 1/2017

Translational Neurodegeneration 1/2017 Go to the issue