Skip to main content
Erschienen in: Current Neurology and Neuroscience Reports 5/2018

01.05.2018 | Movement Disorders (S Fox, Section Editor)

New Imaging Markers for Movement Disorders

verfasst von: Christine Ghadery, Antonio P. Strafella

Erschienen in: Current Neurology and Neuroscience Reports | Ausgabe 5/2018

Einloggen, um Zugang zu erhalten

Abstract

Purpose of Review

For decades, identifying in vivo imaging biomarkers to accurately differentiate between various movement disorders as well as to understand their underlying pathophysiological abnormalities has been the aim of scientific work. Recent advances in multimodal imaging enable the visualization of structural and functional brain changes in these pathological conditions, thus raising the value of imaging techniques as powerful tools to improve sensitivity and specificity of clinical diagnoses. This article reviews well-established and recent developments in imaging markers for movement disorders.

Recent Findings

Whereas several imaging approaches seem to be promising, many modalities are still under development and may not provide decisive answers. Thus, the use of combined imaging modalities as well as the acquisition of methodological consensus in the scientific community may provide more conclusive findings in the future of biomarkers.

Summary

Although a single biomarker has yet not been identified, multiple markers derived from different imaging modalities may represent the right approach.
Literatur
1.
Zurück zum Zitat Dexter DT, Jenner P. Parkinson disease: from pathology to molecular disease mechanisms. Free Radic Biol Med. 2013;62:132–44.PubMedCrossRef Dexter DT, Jenner P. Parkinson disease: from pathology to molecular disease mechanisms. Free Radic Biol Med. 2013;62:132–44.PubMedCrossRef
2.
Zurück zum Zitat Saeed U, Compagnone J, Aviv RI, Strafella AP, Black SE, Lang AE, Masellis M Imaging biomarkers in Parkinson’s disease and Parkinsonian syndromes: current and emerging concepts. Transl Neurodegener 2017 mar 8;6:8-017-0076-6. eCollection 2017. Saeed U, Compagnone J, Aviv RI, Strafella AP, Black SE, Lang AE, Masellis M Imaging biomarkers in Parkinson’s disease and Parkinsonian syndromes: current and emerging concepts. Transl Neurodegener 2017 mar 8;6:8-017-0076-6. eCollection 2017.
3.
Zurück zum Zitat Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55(3):181–4.PubMedPubMedCentralCrossRef Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55(3):181–4.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat de la Fuente-Fernandez R, Schulzer M, Kuramoto L, Cragg J, Ramachandiran N, Au WL, et al. Age-specific progression of nigrostriatal dysfunction in Parkinson’s disease. Ann Neurol. 2011;69(5):803–10.PubMedCrossRef de la Fuente-Fernandez R, Schulzer M, Kuramoto L, Cragg J, Ramachandiran N, Au WL, et al. Age-specific progression of nigrostriatal dysfunction in Parkinson’s disease. Ann Neurol. 2011;69(5):803–10.PubMedCrossRef
5.
Zurück zum Zitat Fernandes Rde C, Berg D. Parenchymal imaging in movement disorders. Front Neurol Neurosci. 2015;36:71–82.PubMedCrossRef Fernandes Rde C, Berg D. Parenchymal imaging in movement disorders. Front Neurol Neurosci. 2015;36:71–82.PubMedCrossRef
6.
Zurück zum Zitat Berg D, Roggendorf W, Schroder 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(6):999–1005.PubMedCrossRef Berg D, Roggendorf W, Schroder 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(6):999–1005.PubMedCrossRef
7.
Zurück zum Zitat 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(4):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(4):448–54.PubMedCrossRef
8.
Zurück zum Zitat Walter U, Dressler D, Wolters A, Probst T, Grossmann A, Benecke R. Sonographic discrimination of corticobasal degeneration vs progressive supranuclear palsy. Neurology. 2004;63(3):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(3):504–9.PubMedCrossRef
9.
Zurück zum Zitat Sadowski K, Serafin-Krol M, Szlachta K, Friedman A. Basal ganglia echogenicity in tauopathies. J Neural Transm (Vienna). 2015;122(6):863–5.CrossRef Sadowski K, Serafin-Krol M, Szlachta K, Friedman A. Basal ganglia echogenicity in tauopathies. J Neural Transm (Vienna). 2015;122(6):863–5.CrossRef
10.
Zurück zum Zitat Maskova J, Skoloudik D, Burgetova A, Fiala O, Bruha R, Zahorakova D, et al. Comparison of transcranial sonography-magnetic resonance fusion imaging in Wilson’s and early-onset Parkinson’s diseases. Parkinsonism Relat Disord. 2016;28:87–93.PubMedCrossRef Maskova J, Skoloudik D, Burgetova A, Fiala O, Bruha R, Zahorakova D, et al. Comparison of transcranial sonography-magnetic resonance fusion imaging in Wilson’s and early-onset Parkinson’s diseases. Parkinsonism Relat Disord. 2016;28:87–93.PubMedCrossRef
11.
Zurück zum Zitat Bor-Seng-Shu E, Pedroso JL, Felicio AC, de Andrade DC, Teixeira MJ, Braga-Neto P, et al. Substantia nigra echogenicity and imaging of striatal dopamine transporters in Parkinson’s disease: a cross-sectional study. Parkinsonism Relat Disord. 2014;20(5):477–81.PubMedCrossRef Bor-Seng-Shu E, Pedroso JL, Felicio AC, de Andrade DC, Teixeira MJ, Braga-Neto P, et al. Substantia nigra echogenicity and imaging of striatal dopamine transporters in Parkinson’s disease: a cross-sectional study. Parkinsonism Relat Disord. 2014;20(5):477–81.PubMedCrossRef
12.
Zurück zum Zitat Tunc S, Graf J, Tadic V, Bruggemann N, Schmidt A, Al-Khaled M, et al. A population-based study on combined markers for early Parkinson’s disease. Mov Disord. 2015;30(4):531–7.PubMedCrossRef Tunc S, Graf J, Tadic V, Bruggemann N, Schmidt A, Al-Khaled M, et al. A population-based study on combined markers for early Parkinson’s disease. Mov Disord. 2015;30(4):531–7.PubMedCrossRef
13.
Zurück zum Zitat Fernandes R, Rosso A, Vincent M, et al. Transcranial sonography of substantia nigra: computer-evaluated echogenicity. Mov Disord. 2012;27(suppl 1):S235. Fernandes R, Rosso A, Vincent M, et al. Transcranial sonography of substantia nigra: computer-evaluated echogenicity. Mov Disord. 2012;27(suppl 1):S235.
14.
Zurück zum Zitat Lehericy S, Vaillancourt DE, Seppi K, Monchi O, Rektorova I, Antonini A, et al. The role of high-field magnetic resonance imaging in parkinsonian disorders: pushing the boundaries forward. Mov Disord. 2017;32(4):510–25.PubMedCrossRef Lehericy S, Vaillancourt DE, Seppi K, Monchi O, Rektorova I, Antonini A, et al. The role of high-field magnetic resonance imaging in parkinsonian disorders: pushing the boundaries forward. Mov Disord. 2017;32(4):510–25.PubMedCrossRef
15.
Zurück zum Zitat Summerfield C, Junque C, Tolosa E, Salgado-Pineda P, Gomez-Anson B, Marti MJ, et al. Structural brain changes in Parkinson disease with dementia: a voxel-based morphometry study. Arch Neurol. 2005;62(2):281–5.PubMedCrossRef Summerfield C, Junque C, Tolosa E, Salgado-Pineda P, Gomez-Anson B, Marti MJ, et al. Structural brain changes in Parkinson disease with dementia: a voxel-based morphometry study. Arch Neurol. 2005;62(2):281–5.PubMedCrossRef
16.
Zurück zum Zitat 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(Pt 4):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(Pt 4):791–800.PubMedCrossRef
17.
Zurück zum Zitat Brenneis C, Seppi K, Schocke MF, Muller J, Luginger E, Bosch S, et al. Voxel-based morphometry detects cortical atrophy in the Parkinson variant of multiple system atrophy. Mov Disord. 2003;18(10):1132–8.PubMedCrossRef Brenneis C, Seppi K, Schocke MF, Muller J, Luginger E, Bosch S, et al. Voxel-based morphometry detects cortical atrophy in the Parkinson variant of multiple system atrophy. Mov Disord. 2003;18(10):1132–8.PubMedCrossRef
18.
Zurück zum Zitat 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(2):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(2):663–9.PubMedCrossRef
19.
Zurück zum Zitat 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(5):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(5):517–23.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat 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):17. -9158-1-17 PubMedPubMedCentralCrossRef 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):17. -9158-1-17 PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Tinaz S, Courtney MG, Stern CE. Focal cortical and subcortical atrophy in early Parkinson’s disease. Mov Disord. 2011;26(3):436–41.PubMedCrossRef Tinaz S, Courtney MG, Stern CE. Focal cortical and subcortical atrophy in early Parkinson’s disease. Mov Disord. 2011;26(3):436–41.PubMedCrossRef
22.
Zurück zum Zitat 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(8):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(8):873–7.PubMedCrossRef
23.
Zurück zum Zitat 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(3):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(3):389–402.PubMedCrossRef
24.
Zurück zum Zitat Kollensperger M, Wenning GK. Assessing disease progression with MRI in atypical parkinsonian disorders. Mov Disord. 2009;24(Suppl 2):S699–702.PubMedCrossRef Kollensperger M, Wenning GK. Assessing disease progression with MRI in atypical parkinsonian disorders. Mov Disord. 2009;24(Suppl 2):S699–702.PubMedCrossRef
25.
Zurück zum Zitat Minnerop M, Specht K, Ruhlmann J, Schimke N, Abele M, Weyer A, et al. Voxel-based morphometry and voxel-based relaxometry in multiple system atrophy-a comparison between clinical subtypes and correlations with clinical parameters. NeuroImage. 2007;36(4):1086–95.PubMedCrossRef Minnerop M, Specht K, Ruhlmann J, Schimke N, Abele M, Weyer A, et al. Voxel-based morphometry and voxel-based relaxometry in multiple system atrophy-a comparison between clinical subtypes and correlations with clinical parameters. NeuroImage. 2007;36(4):1086–95.PubMedCrossRef
26.
Zurück zum Zitat 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(Pt 4):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(Pt 4):1040–9.PubMedCrossRef
27.
Zurück zum Zitat Nicoletti G, Fera F, Condino F, Auteri W, Gallo O, Pugliese P, et al. MR imaging of middle cerebellar peduncle width: differentiation of multiple system atrophy from Parkinson disease. Radiology. 2006;239(3):825–30.PubMedCrossRef Nicoletti G, Fera F, Condino F, Auteri W, Gallo O, Pugliese P, et al. MR imaging of middle cerebellar peduncle width: differentiation of multiple system atrophy from Parkinson disease. Radiology. 2006;239(3):825–30.PubMedCrossRef
28.
Zurück zum Zitat 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(1):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(1):214–21.PubMedCrossRef
29.
Zurück zum Zitat • 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(3):564–70. This study reports reduced volumes of the olfactory bulb in patients with idiopathic Parkinson's disease compared to multiple system atrophy. 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(3):564–70. This study reports reduced volumes of the olfactory bulb in patients with idiopathic Parkinson's disease compared to multiple system atrophy. PubMedCrossRef
30.
Zurück zum Zitat Pietracupa S, Martin-Bastida A, Piccini P. Iron metabolism and its detection through MRI in parkinsonian disorders: a systematic review. Neurol Sci. 2017;38:2095–101.PubMedCrossRef Pietracupa S, Martin-Bastida A, Piccini P. Iron metabolism and its detection through MRI in parkinsonian disorders: a systematic review. Neurol Sci. 2017;38:2095–101.PubMedCrossRef
31.
Zurück zum Zitat Beyer MK, Larsen JP, Aarsland D. Gray matter atrophy in Parkinson disease with dementia and dementia with Lewy bodies. Neurology. 2007;69(8):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(8):747–54.PubMedCrossRef
32.
Zurück zum Zitat 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(6):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(6):727–34.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat 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. AJNR Am J Neuroradiol. 2014;35(12):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. AJNR Am J Neuroradiol. 2014;35(12):2257–64.PubMedCrossRef
34.
Zurück zum Zitat Beyer MK, Aarsland D, Greve OJ, Larsen JP. Visual rating of white matter hyperintensities in Parkinson’s disease. Mov Disord. 2006;21(2):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(2):223–9.PubMedCrossRef
35.
Zurück zum Zitat Fanciulli A, Wenning GK. Multiple-system atrophy. N Engl J Med. 2015;372(14):1375–6.PubMed Fanciulli A, Wenning GK. Multiple-system atrophy. N Engl J Med. 2015;372(14):1375–6.PubMed
36.
Zurück zum Zitat 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(4):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(4):457–63.PubMedCrossRef
37.
Zurück zum Zitat 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(1):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(1):81–6.PubMedCrossRef
38.
Zurück zum Zitat 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(22):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(22):2775–81.PubMedCrossRef
39.
Zurück zum Zitat 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 Feb;29(2):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 Feb;29(2):280–9.PubMedCrossRef
40.
Zurück zum Zitat Groschel 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(2):714–24.PubMedCrossRef Groschel 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(2):714–24.PubMedCrossRef
41.
Zurück zum Zitat 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(1):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(1):34–9.PubMedCrossRef
42.
Zurück zum Zitat Lee SE, Rabinovici GD, Mayo MC, Wilson SM, Seeley WW, DeArmond SJ, et al. Clinicopathological correlations in corticobasal degeneration. Ann Neurol. 2011;70(2):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(2):327–40.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat • 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(7):1251–3. This study replicated findings on the midbrain-to-pons ratio which demonstrated high specificity and sensitivity for the diagnosis of progressive supranuclear palsy. 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(7):1251–3. This study replicated findings on the midbrain-to-pons ratio which demonstrated high specificity and sensitivity for the diagnosis of progressive supranuclear palsy. PubMedCrossRef
44.
Zurück zum Zitat 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(12):1881–4.PubMedCrossRef 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(12):1881–4.PubMedCrossRef
45.
Zurück zum Zitat 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(12):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(12):1863–72.PubMedCrossRef
46.
Zurück zum Zitat 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(Pt 1):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(Pt 1):170–80.PubMedCrossRef
47.
Zurück zum Zitat • Du G, Liu T, Lewis MM, Kong L, Wang Y, Connor J, et al. Quantitative susceptibility mapping of the midbrain in Parkinson’s disease. Mov Disord. 2016;31(3):317–24. The authors of this study revealed that quantitative susceptibility mapping may be a superior imaging biomarker to R2* for estimating brain iron levels in PD. PubMedCrossRef • Du G, Liu T, Lewis MM, Kong L, Wang Y, Connor J, et al. Quantitative susceptibility mapping of the midbrain in Parkinson’s disease. Mov Disord. 2016;31(3):317–24. The authors of this study revealed that quantitative susceptibility mapping may be a superior imaging biomarker to R2* for estimating brain iron levels in PD. PubMedCrossRef
48.
Zurück zum Zitat Reimao S, Ferreira S, Nunes RG, Pita Lobo P, Neutel D, Abreu D, et al. Magnetic resonance correlation of iron content with neuromelanin in the substantia nigra of early-stage Parkinson’s disease. Eur J Neurol. 2016;23(2):368–74.PubMedCrossRef Reimao S, Ferreira S, Nunes RG, Pita Lobo P, Neutel D, Abreu D, et al. Magnetic resonance correlation of iron content with neuromelanin in the substantia nigra of early-stage Parkinson’s disease. Eur J Neurol. 2016;23(2):368–74.PubMedCrossRef
49.
Zurück zum Zitat Dexter DT, Jenner P, Schapira AH, Marsden CD. Alterations in levels of iron, ferritin, and other trace metals in neurodegenerative diseases affecting the basal ganglia. The Royal Kings and Queens Parkinson's Disease Research Group. Ann Neurol. 1992;32(Suppl):S94–100.PubMedCrossRef Dexter DT, Jenner P, Schapira AH, Marsden CD. Alterations in levels of iron, ferritin, and other trace metals in neurodegenerative diseases affecting the basal ganglia. The Royal Kings and Queens Parkinson's Disease Research Group. Ann Neurol. 1992;32(Suppl):S94–100.PubMedCrossRef
50.
Zurück zum Zitat Boelmans K, Holst B, Hackius M, Finsterbusch J, Gerloff C, Fiehler J, et al. Brain iron deposition fingerprints in Parkinson’s disease and progressive supranuclear palsy. Mov Disord. 2012;27(3):421–7.PubMedCrossRef Boelmans K, Holst B, Hackius M, Finsterbusch J, Gerloff C, Fiehler J, et al. Brain iron deposition fingerprints in Parkinson’s disease and progressive supranuclear palsy. Mov Disord. 2012;27(3):421–7.PubMedCrossRef
51.
Zurück zum Zitat Sakurai K, Imabayashi E, Tokumaru AM, Ito K, Shimoji K, Nakagawa M, et al. Volume of interest analysis of spatially normalized PRESTO imaging to differentiate between Parkinson disease and atypical Parkinsonian syndrome. Magn Reson Med Sci. 2017;16(1):16–22.PubMedCrossRef Sakurai K, Imabayashi E, Tokumaru AM, Ito K, Shimoji K, Nakagawa M, et al. Volume of interest analysis of spatially normalized PRESTO imaging to differentiate between Parkinson disease and atypical Parkinsonian syndrome. Magn Reson Med Sci. 2017;16(1):16–22.PubMedCrossRef
52.
Zurück zum Zitat 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(6):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(6):534–40.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Mahlknecht P, Krismer F, Poewe W, Seppi K. Meta-analysis of dorsolateral nigral hyperintensity on magnetic resonance imaging as a marker for Parkinson’s disease. Mov Disord. 2017;32(4):619–23.PubMedCrossRef Mahlknecht P, Krismer F, Poewe W, Seppi K. Meta-analysis of dorsolateral nigral hyperintensity on magnetic resonance imaging as a marker for Parkinson’s disease. Mov Disord. 2017;32(4):619–23.PubMedCrossRef
54.
Zurück zum Zitat •• De Marzi R, Seppi K, Hogl B, Muller C, Scherfler C, Stefani A, et al. Loss of dorsolateral nigral hyperintensity on 3.0 tesla susceptibility-weighted imaging in idiopathic rapid eye movement sleep behavior disorder. Ann Neurol. 2016;79(6):1026–30. This study identified the absence of dorsolateral nigral hyperintensity on high-field susceptibility-weighted imaging as a potential biomarker for prodromal degenerative parkinsonism in idiopathic rapid eye movement sleep behavior disorder. PubMedCrossRef •• De Marzi R, Seppi K, Hogl B, Muller C, Scherfler C, Stefani A, et al. Loss of dorsolateral nigral hyperintensity on 3.0 tesla susceptibility-weighted imaging in idiopathic rapid eye movement sleep behavior disorder. Ann Neurol. 2016;79(6):1026–30. This study identified the absence of dorsolateral nigral hyperintensity on high-field susceptibility-weighted imaging as a potential biomarker for prodromal degenerative parkinsonism in idiopathic rapid eye movement sleep behavior disorder. PubMedCrossRef
55.
Zurück zum Zitat Double KL, Gerlach M, Schunemann V, Trautwein AX, Zecca L, Gallorini M, et al. Iron-binding characteristics of neuromelanin of the human substantia nigra. Biochem Pharmacol. 2003;66(3):489–94.PubMedCrossRef Double KL, Gerlach M, Schunemann V, Trautwein AX, Zecca L, Gallorini M, et al. Iron-binding characteristics of neuromelanin of the human substantia nigra. Biochem Pharmacol. 2003;66(3):489–94.PubMedCrossRef
56.
Zurück zum Zitat •• Reimao 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(7):953–9. This study revealed that neuromelanin-sensitive MRI techniques can discriminate essential tremor from early-stage tremor-dominant Parkinson's disease. PubMedCrossRef •• Reimao 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(7):953–9. This study revealed that neuromelanin-sensitive MRI techniques can discriminate essential tremor from early-stage tremor-dominant Parkinson's disease. PubMedCrossRef
57.
Zurück zum Zitat Cochrane CJ, Ebmeier KP. Diffusion tensor imaging in parkinsonian syndromes: a systematic review and meta-analysis. Neurology. 2013;80(9):857–64.PubMedPubMedCentralCrossRef Cochrane CJ, Ebmeier KP. Diffusion tensor imaging in parkinsonian syndromes: a systematic review and meta-analysis. Neurology. 2013;80(9):857–64.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Schocke MF, Seppi K, Esterhammer R, Kremser C, Mair KJ, Czermak BV, et al. Trace of diffusion tensor differentiates the Parkinson variant of multiple system atrophy and Parkinson’s disease. NeuroImage. 2004;21(4):1443–51.PubMedCrossRef Schocke MF, Seppi K, Esterhammer R, Kremser C, Mair KJ, Czermak BV, et al. Trace of diffusion tensor differentiates the Parkinson variant of multiple system atrophy and Parkinson’s disease. NeuroImage. 2004;21(4):1443–51.PubMedCrossRef
59.
Zurück zum Zitat 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(3):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(3):380–7.PubMedCrossRef
60.
Zurück zum Zitat •• Ofori E, Krismer F, Burciu RG, Pasternak O, McCracken JL, Lewis MM, et al. Free water improves detection of changes in the substantia nigra in parkinsonism: a multisite study. Mov Disord 2017 Jul 17. This multisite study used single- and bi-tensor models of diffusion magnetic resonance imaging to evaluate changes in the substantia nigra in PD, MSA, and PSP. •• Ofori E, Krismer F, Burciu RG, Pasternak O, McCracken JL, Lewis MM, et al. Free water improves detection of changes in the substantia nigra in parkinsonism: a multisite study. Mov Disord 2017 Jul 17. This multisite study used single- and bi-tensor models of diffusion magnetic resonance imaging to evaluate changes in the substantia nigra in PD, MSA, and PSP.
61.
Zurück zum Zitat Ciurleo R, Di Lorenzo G, Bramanti P, Marino S. Magnetic resonance spectroscopy: an in vivo molecular imaging biomarker for Parkinson’s disease? Biomed Res Int 2014;2014:519816, 1, 10. Ciurleo R, Di Lorenzo G, Bramanti P, Marino S. Magnetic resonance spectroscopy: an in vivo molecular imaging biomarker for Parkinson’s disease? Biomed Res Int 2014;2014:519816, 1, 10.
62.
Zurück zum Zitat Kim J, Criaud M, Cho S, Cirarda M, Mihaescu A, Coakeley S, et al. Abnormal intrinsic brain functional network dynamics in Parkinson’s disease. Brain : a journal of neurology 2017;accepted, 140, 2955, 2967. Kim J, Criaud M, Cho S, Cirarda M, Mihaescu A, Coakeley S, et al. Abnormal intrinsic brain functional network dynamics in Parkinson’s disease. Brain : a journal of neurology 2017;accepted, 140, 2955, 2967.
63.
Zurück zum Zitat Brooks DJ. Technology insight: imaging neurodegeneration in Parkinson’s disease. Nat Clin Pract Neurol. 2008;4(5):267–77.PubMedCrossRef Brooks DJ. Technology insight: imaging neurodegeneration in Parkinson’s disease. Nat Clin Pract Neurol. 2008;4(5):267–77.PubMedCrossRef
64.
Zurück zum Zitat Broussolle E, Dentresangle C, Landais P, Garcia-Larrea L, Pollak P, Croisile B, et al. The relation of putamen and caudate nucleus 18F-Dopa uptake to motor and cognitive performances in Parkinson’s disease. J Neurol Sci. 1999;166(2):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 18F-Dopa uptake to motor and cognitive performances in Parkinson’s disease. J Neurol Sci. 1999;166(2):141–51.PubMedCrossRef
65.
Zurück zum Zitat Otsuka M, Ichiya Y, Hosokawa S, Kuwabara Y, Tahara T, Fukumura T, et al. Striatal blood flow, glucose metabolism and 18F-dopa uptake: difference in Parkinson’s disease and atypical parkinsonism. J Neurol Neurosurg Psychiatry. 1991;54(10):898–904.PubMedPubMedCentralCrossRef Otsuka M, Ichiya Y, Hosokawa S, Kuwabara Y, Tahara T, Fukumura T, et al. Striatal blood flow, glucose metabolism and 18F-dopa uptake: difference in Parkinson’s disease and atypical parkinsonism. J Neurol Neurosurg Psychiatry. 1991;54(10):898–904.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Politis M. Neuroimaging in Parkinson disease: from research setting to clinical practice. Nat Rev Neurol. 2014;10(12):708–22.PubMedCrossRef Politis M. Neuroimaging in Parkinson disease: from research setting to clinical practice. Nat Rev Neurol. 2014;10(12):708–22.PubMedCrossRef
67.
Zurück zum Zitat 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(10):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(10):1724–7.PubMedCrossRef
68.
Zurück zum Zitat Im JH, Chung SJ, Kim JS, 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(1–2):103–9.PubMedCrossRef Im JH, Chung SJ, Kim JS, 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(1–2):103–9.PubMedCrossRef
69.
Zurück zum Zitat 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(6):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(6):919–25.PubMedCrossRef
70.
Zurück zum Zitat 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(4):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(4):907–12.PubMedCrossRef
71.
Zurück zum Zitat 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(4):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(4):493–503.PubMedCrossRef
72.
Zurück zum Zitat 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(7):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(7):1325–9.PubMedCrossRef
73.
Zurück zum Zitat 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(6):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(6):1206–14.PubMedCrossRef
74.
Zurück zum Zitat 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;17(2):303–12.PubMedCrossRef 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;17(2):303–12.PubMedCrossRef
75.
Zurück zum Zitat 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 ( Pt 12)(Pt 12):2187–2195. 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 ( Pt 12)(Pt 12):2187–2195.
76.
Zurück zum Zitat 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. Parkinsonism Relat Disord. 2013;19(2):222–6.PubMedCrossRef 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. Parkinsonism Relat Disord. 2013;19(2):222–6.PubMedCrossRef
77.
Zurück zum Zitat 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(8):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(8):1265–73.PubMedCrossRef
78.
Zurück zum Zitat Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Oertel WH, et al. Serotonin neuron loss and nonmotor symptoms continue in Parkinson’s patients treated with dopamine grafts. Sci Transl Med. 2012;4(128):128ra41.PubMedCrossRef Politis M, Wu K, Loane C, Quinn NP, Brooks DJ, Oertel WH, et al. Serotonin neuron loss and nonmotor symptoms continue in Parkinson’s patients treated with dopamine grafts. Sci Transl Med. 2012;4(128):128ra41.PubMedCrossRef
79.
Zurück zum Zitat Roussakis AA, Politis M, Towey D, Piccini P. Serotonin-to-dopamine transporter ratios in Parkinson disease: relevance for dyskinesias. Neurology. 2016;86(12):1152–8.PubMedCrossRef Roussakis AA, Politis M, Towey D, Piccini P. Serotonin-to-dopamine transporter ratios in Parkinson disease: relevance for dyskinesias. Neurology. 2016;86(12):1152–8.PubMedCrossRef
80.
Zurück zum Zitat Politis M, Oertel WH, Wu K, Quinn NP, Pogarell O, Brooks DJ, et al. Graft-induced dyskinesias in Parkinson’s disease: high striatal serotonin/dopamine transporter ratio. Mov Disord. 2011;26(11):1997–2003.PubMedCrossRef Politis M, Oertel WH, Wu K, Quinn NP, Pogarell O, Brooks DJ, et al. Graft-induced dyskinesias in Parkinson’s disease: high striatal serotonin/dopamine transporter ratio. Mov Disord. 2011;26(11):1997–2003.PubMedCrossRef
81.
Zurück zum Zitat Niccolini F, Foltynie T, Reis Marques T, Muhlert N, Tziortzi AC, Searle GE, et al. Loss of phosphodiesterase 10A expression is associated with progression and severity in Parkinson’s disease. Brain. 2015;138(Pt 10):3003–15.PubMedCrossRef Niccolini F, Foltynie T, Reis Marques T, Muhlert N, Tziortzi AC, Searle GE, et al. Loss of phosphodiesterase 10A expression is associated with progression and severity in Parkinson’s disease. Brain. 2015;138(Pt 10):3003–15.PubMedCrossRef
82.
Zurück zum Zitat 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. 2013;47(1):44–51.PubMedCrossRef 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. 2013;47(1):44–51.PubMedCrossRef
83.
Zurück zum Zitat Fazio P, Svenningsson P, Forsberg A, Jonsson EG, Amini N, Nakao R, et al. Quantitative analysis of (1)(8)F-(E)-N-(3-Iodoprop-2-Enyl)-2beta-Carbofluoroethoxy-3beta-(4′-methyl-phenyl) nortropane binding to the dopamine transporter in Parkinson disease. J Nucl Med. 2015;56(5):714–20.PubMedCrossRef Fazio P, Svenningsson P, Forsberg A, Jonsson EG, Amini N, Nakao R, et al. Quantitative analysis of (1)(8)F-(E)-N-(3-Iodoprop-2-Enyl)-2beta-Carbofluoroethoxy-3beta-(4′-methyl-phenyl) nortropane binding to the dopamine transporter in Parkinson disease. J Nucl Med. 2015;56(5):714–20.PubMedCrossRef
84.
Zurück zum Zitat 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(8):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(8):965–73.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat 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(15):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(15):2516–23.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Donaghy P, Thomas AJ, O'Brien JT. Amyloid PET imaging in Lewy body disorders. Am J Geriatr Psychiatry. 2015;23(1):23–37.PubMedCrossRef Donaghy P, Thomas AJ, O'Brien JT. Amyloid PET imaging in Lewy body disorders. Am J Geriatr Psychiatry. 2015;23(1):23–37.PubMedCrossRef
87.
Zurück zum Zitat Kepe V, Bordelon Y, Boxer A, Huang SC, Liu J, Thiede FC, et al. PET imaging of neuropathology in tauopathies: progressive supranuclear palsy. J Alzheimers Dis. 2013;36(1):145–53.PubMedPubMedCentralCrossRef Kepe V, Bordelon Y, Boxer A, Huang SC, Liu J, Thiede FC, et al. PET imaging of neuropathology in tauopathies: progressive supranuclear palsy. J Alzheimers Dis. 2013;36(1):145–53.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat 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. 2017;32(1):134–40.PubMedCrossRef 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. 2017;32(1):134–40.PubMedCrossRef
89.
Zurück zum Zitat Hammes J, Bischof GN, Giehl K, Faber J, Drzezga A, Klockgether T, et al. Elevated in vivo [18F]-AV-1451 uptake in a patient with progressive supranuclear palsy. Mov Disord. 2017;32(1):170–1.PubMedCrossRef Hammes J, Bischof GN, Giehl K, Faber J, Drzezga A, Klockgether T, et al. Elevated in vivo [18F]-AV-1451 uptake in a patient with progressive supranuclear palsy. Mov Disord. 2017;32(1):170–1.PubMedCrossRef
90.
Zurück zum Zitat •• Coakeley S, Cho SS, Koshimori Y, Rusjan P, Ghadery C, Kim J, et al. [18F]AV-1451 binding to neuromelanin in the substantia nigra in PD and PSP. Brain Struct Funct 2017 Sep 7. This study detected that [18F]AV-1451 may be the first PET radiotracer capable of imaging neurodegeneration of the substantia nigra in parkinsonisms. •• Coakeley S, Cho SS, Koshimori Y, Rusjan P, Ghadery C, Kim J, et al. [18F]AV-1451 binding to neuromelanin in the substantia nigra in PD and PSP. Brain Struct Funct 2017 Sep 7. This study detected that [18F]AV-1451 may be the first PET radiotracer capable of imaging neurodegeneration of the substantia nigra in parkinsonisms.
91.
Zurück zum Zitat Coakeley S, Cho SS, Koshimori Y, Rusjan P, Harris M, Ghadery C, et al. Positron emission tomography imaging of tau pathology in progressive supranuclear palsy. J Cereb Blood Flow Metab 2016 Jan 01:271678X16683695. Coakeley S, Cho SS, Koshimori Y, Rusjan P, Harris M, Ghadery C, et al. Positron emission tomography imaging of tau pathology in progressive supranuclear palsy. J Cereb Blood Flow Metab 2016 Jan 01:271678X16683695.
92.
Zurück zum Zitat 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 Sep 9;61(5):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 Sep 9;61(5):686–9.PubMedCrossRef
93.
Zurück zum Zitat Kobylecki C, Counsell SJ, Cabanel N, Wachter T, Turkheimer FE, Eggert K, et al. Diffusion-weighted imaging and its relationship to microglial activation in parkinsonian syndromes. Parkinsonism Relat Disord. 2013;19(5):527–32.PubMedCrossRef Kobylecki C, Counsell SJ, Cabanel N, Wachter T, Turkheimer FE, Eggert K, et al. Diffusion-weighted imaging and its relationship to microglial activation in parkinsonian syndromes. Parkinsonism Relat Disord. 2013;19(5):527–32.PubMedCrossRef
94.
Zurück zum Zitat 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(10):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(10):1221–6.PubMedCrossRef
95.
Zurück zum Zitat Gerhard A, Trender-Gerhard I, Turkheimer F, Quinn NP, Bhatia KP, Brooks DJ. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in progressive supranuclear palsy. Mov Disord. 2006;21(1):89–93.PubMedCrossRef Gerhard A, Trender-Gerhard I, Turkheimer F, Quinn NP, Bhatia KP, Brooks DJ. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in progressive supranuclear palsy. Mov Disord. 2006;21(1):89–93.PubMedCrossRef
96.
Zurück zum Zitat Terada T, Yokokura M, Yoshikawa E, Futatsubashi M, Kono S, Konishi T, et al. Extrastriatal spreading of microglial activation in Parkinson’s disease: a positron emission tomography study. Ann Nucl Med. 2016;30:579–87.PubMedCrossRef Terada T, Yokokura M, Yoshikawa E, Futatsubashi M, Kono S, Konishi T, et al. Extrastriatal spreading of microglial activation in Parkinson’s disease: a positron emission tomography study. Ann Nucl Med. 2016;30:579–87.PubMedCrossRef
97.
Zurück zum Zitat 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(6):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(6):938–49.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat 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. Alzheimers Dement. 2015;11(6):608–21.e7.PubMedCrossRef 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. Alzheimers Dement. 2015;11(6):608–21.e7.PubMedCrossRef
99.
Zurück zum Zitat Iannaccone S, Cerami C, Alessio M, Garibotto V, Panzacchi A, Olivieri S, et al. In vivo microglia activation in very early dementia with Lewy bodies, comparison with Parkinson’s disease. Parkinsonism Relat Disord. 2013;19(1):47–52.PubMedCrossRef Iannaccone S, Cerami C, Alessio M, Garibotto V, Panzacchi A, Olivieri S, et al. In vivo microglia activation in very early dementia with Lewy bodies, comparison with Parkinson’s disease. Parkinsonism Relat Disord. 2013;19(1):47–52.PubMedCrossRef
100.
Zurück zum Zitat Ghadery C, Koshimori Y, Coakeley S, Harris M, Rusjan P, Kim J, et al. Microglial activation in Parkinson’s disease using [18F]-FEPPA. J Neuroinflammation. 2017;14(1):8. -016-0778-1 PubMedPubMedCentralCrossRef Ghadery C, Koshimori Y, Coakeley S, Harris M, Rusjan P, Kim J, et al. Microglial activation in Parkinson’s disease using [18F]-FEPPA. J Neuroinflammation. 2017;14(1):8. -016-0778-1 PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Koshimori Y, Ko JH, Mizrahi R, Rusjan P, Mabrouk R, Jacobs MF, et al. Imaging striatal microglial activation in patients with Parkinson’s disease. PLoS One. 2015;10(9):e0138721.PubMedPubMedCentralCrossRef Koshimori Y, Ko JH, Mizrahi R, Rusjan P, Mabrouk R, Jacobs MF, et al. Imaging striatal microglial activation in patients with Parkinson’s disease. PLoS One. 2015;10(9):e0138721.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Botha H, Whitwell JL, Madhaven A, Senjem ML, Lowe V, Josephs KA. The pimple sign of progressive supranuclear palsy syndrome. Parkinsonism Relat Disord. 2014;20(2):180–5.PubMedCrossRef Botha H, Whitwell JL, Madhaven A, Senjem ML, Lowe V, Josephs KA. The pimple sign of progressive supranuclear palsy syndrome. Parkinsonism Relat Disord. 2014;20(2):180–5.PubMedCrossRef
103.
Zurück zum Zitat 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(Pt 11):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(Pt 11):3036–46.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat 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 Mar;55(4):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 Mar;55(4):483–92.PubMedCrossRef
105.
Zurück zum Zitat •• Holtbernd F, Ma Y, Peng S, Schwartz F, Timmermann L, Kracht L, et al. Dopaminergic correlates of metabolic network activity in Parkinson’s disease. Hum Brain Mapp. 2015;36(9):3575–85. This study found that Parkinson’s disease motor- and cognition-related metabolic patterns correlated significantly with PET indices of presynaptic dopaminergic functioning in patients with Parkinson's disease. PubMedCrossRef •• Holtbernd F, Ma Y, Peng S, Schwartz F, Timmermann L, Kracht L, et al. Dopaminergic correlates of metabolic network activity in Parkinson’s disease. Hum Brain Mapp. 2015;36(9):3575–85. This study found that Parkinson’s disease motor- and cognition-related metabolic patterns correlated significantly with PET indices of presynaptic dopaminergic functioning in patients with Parkinson's disease. PubMedCrossRef
106.
Zurück zum Zitat Coakeley S, Strafella AP. Imaging tau pathology in Parkinsonisms. NPJ Parkinsons Dis 2017 29;3:22-017-0023-3. eCollection 2017. Coakeley S, Strafella AP. Imaging tau pathology in Parkinsonisms. NPJ Parkinsons Dis 2017 29;3:22-017-0023-3. eCollection 2017.
107.
Zurück zum Zitat Rupprecht R, Papadopoulos V, Rammes G, Baghai TC, Fan J, Akula N, et al. Translocator protein (18 kDa) (TSPO) as a therapeutic target for neurological and psychiatric disorders. Nat Rev Drug Discov. 2010;9(12):971–88.PubMedCrossRef Rupprecht R, Papadopoulos V, Rammes G, Baghai TC, Fan J, Akula N, et al. Translocator protein (18 kDa) (TSPO) as a therapeutic target for neurological and psychiatric disorders. Nat Rev Drug Discov. 2010;9(12):971–88.PubMedCrossRef
108.
Zurück zum Zitat Koshimori Y, Ko JH, Mizrahi R, Rusjan P, Mabrouk R, Jacobs MF, et al. Imaging striatal microglial activation in patients with Parkinson’s disease. PLoS One. 2015;10(9):e0138721.PubMedPubMedCentralCrossRef Koshimori Y, Ko JH, Mizrahi R, Rusjan P, Mabrouk R, Jacobs MF, et al. Imaging striatal microglial activation in patients with Parkinson’s disease. PLoS One. 2015;10(9):e0138721.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Gjerloff T, Fedorova T, Knudsen K, Munk OL, Nahimi A, Jacobsen S, et al. Imaging acetylcholinesterase density in peripheral organs in Parkinson’s disease with 11C-donepezil PET. Brain. 2015;138(Pt 3):653–63.PubMedCrossRef Gjerloff T, Fedorova T, Knudsen K, Munk OL, Nahimi A, Jacobsen S, et al. Imaging acetylcholinesterase density in peripheral organs in Parkinson’s disease with 11C-donepezil PET. Brain. 2015;138(Pt 3):653–63.PubMedCrossRef
Metadaten
Titel
New Imaging Markers for Movement Disorders
verfasst von
Christine Ghadery
Antonio P. Strafella
Publikationsdatum
01.05.2018
Verlag
Springer US
Erschienen in
Current Neurology and Neuroscience Reports / Ausgabe 5/2018
Print ISSN: 1528-4042
Elektronische ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-018-0830-x

Weitere Artikel der Ausgabe 5/2018

Current Neurology and Neuroscience Reports 5/2018 Zur Ausgabe

Movement Disorders (S Fox, Section Editor)

Managing Gait, Balance, and Posture in Parkinson’s Disease

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

„Restriktion auf vier Wochen Therapie bei Schlaflosigkeit ist absurd!“

06.05.2024 Insomnie Nachrichten

Chronische Insomnie als eigenständiges Krankheitsbild ernst nehmen und adäquat nach dem aktuellen Forschungsstand behandeln: Das forderte der Schlafmediziner Dr. Dieter Kunz von der Berliner Charité beim Praxis Update.

Stuhltransfusion könnte Fortschreiten von Parkinson-Symptomen bremsen

03.05.2024 Parkinson-Krankheit Nachrichten

Kann eine frühzeitige Stuhltransplantation das Fortschreiten von Parkinson-Symptomen verlangsamen? Die Ergebnisse einer randomisierten Phase-2-Studie scheinen dafür zu sprechen.

Frühe Tranexamsäure-Therapie nützt wenig bei Hirnblutungen

02.05.2024 Hirnblutung Nachrichten

Erhalten Personen mit einer spontanen Hirnblutung innerhalb von zwei Stunden nach Symptombeginn eine Tranexamsäure-Therapie, kann dies weder die Hämatomexpansion eindämmen noch die Mortalität senken.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders scheint das auf weibliche Kranke zuzutreffen, wie eine Studie zeigt.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.