Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 9/2013

01-09-2013 | Neuroimaging (DJ Brooks)

Cholinergic Dysfunction in Parkinson’s Disease

Authors: Martijn L. T. M. Müller, Nicolaas I. Bohnen

Published in: Current Neurology and Neuroscience Reports | Issue 9/2013

Login to get access

Abstract

There is increasing interest in the clinical effects of cholinergic basal forebrain and tegmental pedunculopontine complex (PPN) projection degeneration in Parkinson’s disease (PD). Recent evidence supports an expanded role beyond cognitive impairment, including effects on olfaction, mood, REM sleep behavior disorder, and motor functions. Cholinergic denervation is variable in PD without dementia and may contribute to clinical symptom heterogeneity. Early in vivo imaging evidence that impaired cholinergic integrity of the PPN associates with frequent falling in PD is now confirmed by human post-mortem evidence. Brainstem cholinergic lesioning studies in primates confirm the role of the PPN in mobility impairment. Degeneration of basal forebrain cholinergic projections correlates with decreased walking speed. Cumulatively, these findings provide evidence for a new paradigm to explain dopamine-resistant features of mobility impairments in PD. Recognition of the increased clinical role of cholinergic system degeneration may motivate new research to expand indications for cholinergic therapy in PD.
Literature
1.
go back to reference Langston JW. The Parkinson’s complex: parkinsonism is just the tip of the iceberg. Ann Neurol. 2006;59:591–6.PubMedCrossRef Langston JW. The Parkinson’s complex: parkinsonism is just the tip of the iceberg. Ann Neurol. 2006;59:591–6.PubMedCrossRef
2.
go back to reference Braak H, Del Tredici K, Rub U, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.PubMedCrossRef Braak H, Del Tredici K, Rub U, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.PubMedCrossRef
3.
go back to reference Bohnen NI, Kaufer DI, Ivanco LS, et al. Cortical cholinergic function is more severely affected in parkinsonian dementia than in Alzheimer disease: an in vivo positron emission tomographic study. Arch Neurol. 2003;60:1745–8.PubMedCrossRef Bohnen NI, Kaufer DI, Ivanco LS, et al. Cortical cholinergic function is more severely affected in parkinsonian dementia than in Alzheimer disease: an in vivo positron emission tomographic study. Arch Neurol. 2003;60:1745–8.PubMedCrossRef
4.
go back to reference Bohnen NI, Albin RL. The cholinergic system and Parkinson disease. Behav Brain Res. 2011;221:564–73.PubMedCrossRef Bohnen NI, Albin RL. The cholinergic system and Parkinson disease. Behav Brain Res. 2011;221:564–73.PubMedCrossRef
5.
go back to reference Mesulam MM, Geula C. Nucleus basalis (Ch4) and cortical cholinergic innervation in the human brain: observations based on the distribution of acetylcholinesterase and choline acetyltransferase. J Comp Neurol. 1988;275:216–40.PubMedCrossRef Mesulam MM, Geula C. Nucleus basalis (Ch4) and cortical cholinergic innervation in the human brain: observations based on the distribution of acetylcholinesterase and choline acetyltransferase. J Comp Neurol. 1988;275:216–40.PubMedCrossRef
6.
go back to reference Heckers S, Geula C, Mesulam MM. Cholinergic innervation of the human thalamus: dual origin and differential nuclear distribution. J Comp Neurol. 1992;325:68–82.PubMedCrossRef Heckers S, Geula C, Mesulam MM. Cholinergic innervation of the human thalamus: dual origin and differential nuclear distribution. J Comp Neurol. 1992;325:68–82.PubMedCrossRef
7.
go back to reference de Lacalle S, Hersh LB, Saper CB. Cholinergic innervation of the human cerebellum. J Comp Neurol. 1993;328:364–76.PubMedCrossRef de Lacalle S, Hersh LB, Saper CB. Cholinergic innervation of the human cerebellum. J Comp Neurol. 1993;328:364–76.PubMedCrossRef
8.
go back to reference Fibiger HC. The organization and some projections of cholinergic neurons of the mammalian forebrain. Brain Res. 1982;257:327–88.PubMed Fibiger HC. The organization and some projections of cholinergic neurons of the mammalian forebrain. Brain Res. 1982;257:327–88.PubMed
9.
go back to reference Lecourtier L, Kelly PH. A conductor hidden in the orchestra? Role of the habenular complex in monoamine transmission and cognition. Neurosci Biobehav Rev. 2007;31:658–72.PubMedCrossRef Lecourtier L, Kelly PH. A conductor hidden in the orchestra? Role of the habenular complex in monoamine transmission and cognition. Neurosci Biobehav Rev. 2007;31:658–72.PubMedCrossRef
10.
go back to reference Mesulam MM, Mash D, Hersh L, et al. Cholinergic innervation of the human striatum, globus pallidus, subthalamic nucleus, substantia nigra, and red nucleus. J Comp Neurol. 1992;323:252–68.PubMedCrossRef Mesulam MM, Mash D, Hersh L, et al. Cholinergic innervation of the human striatum, globus pallidus, subthalamic nucleus, substantia nigra, and red nucleus. J Comp Neurol. 1992;323:252–68.PubMedCrossRef
11.
go back to reference Flores CM, Rogers SW, Pabreza LA, et al. A subtype of nicotinic cholinergic receptor in rat brain is composed of α4 and β2 subunits and is up-regulated by chronic nicotine treatment. Mol Pharmacol. 1992;41:31–7.PubMed Flores CM, Rogers SW, Pabreza LA, et al. A subtype of nicotinic cholinergic receptor in rat brain is composed of α4 and β2 subunits and is up-regulated by chronic nicotine treatment. Mol Pharmacol. 1992;41:31–7.PubMed
12.
go back to reference Court J, Clementi F. Distribution of nicotinic subtypes in human brain. Alzheimer Dis Assoc Disord. 1995;9 Suppl 2:6–14.PubMedCrossRef Court J, Clementi F. Distribution of nicotinic subtypes in human brain. Alzheimer Dis Assoc Disord. 1995;9 Suppl 2:6–14.PubMedCrossRef
13.
go back to reference Cortes R, Palacios JM. Muscarinic cholinergic receptor subtypes in the rat brain. I. Quantitative autoradiographic studies. Brain Res. 1986;362:227–38.PubMedCrossRef Cortes R, Palacios JM. Muscarinic cholinergic receptor subtypes in the rat brain. I. Quantitative autoradiographic studies. Brain Res. 1986;362:227–38.PubMedCrossRef
14.
go back to reference Cortes R, Probst A, Tobler HJ, Palacios JM. Muscarinic cholinergic receptor subtypes in the human brain. II. Quantitative autoradiographic studies. Brain Res. 1986;362:239–53.PubMedCrossRef Cortes R, Probst A, Tobler HJ, Palacios JM. Muscarinic cholinergic receptor subtypes in the human brain. II. Quantitative autoradiographic studies. Brain Res. 1986;362:239–53.PubMedCrossRef
15.
go back to reference Mesulam M, Shaw P, Mash D, Weintraub S. Cholinergic nucleus basalis tauopathy emerges early in the aging-MCI-AD continuum. Ann Neurol. 2004;55:815–28.PubMedCrossRef Mesulam M, Shaw P, Mash D, Weintraub S. Cholinergic nucleus basalis tauopathy emerges early in the aging-MCI-AD continuum. Ann Neurol. 2004;55:815–28.PubMedCrossRef
16.
go back to reference Kotagal V, Muller ML, Kaufer DI, et al. Thalamic cholinergic innervation is spared in Alzheimer disease compared with parkinsonian disorders. Neurosci Lett. 2012;514:169–72.PubMedCrossRef Kotagal V, Muller ML, Kaufer DI, et al. Thalamic cholinergic innervation is spared in Alzheimer disease compared with parkinsonian disorders. Neurosci Lett. 2012;514:169–72.PubMedCrossRef
17.
go back to reference Davis KL, Mohs RC, Marin D, et al. Cholinergic markers in elderly patients with early signs of Alzheimer disease. JAMA. 1999;281:1401–6.PubMedCrossRef Davis KL, Mohs RC, Marin D, et al. Cholinergic markers in elderly patients with early signs of Alzheimer disease. JAMA. 1999;281:1401–6.PubMedCrossRef
18.
go back to reference DeKosky ST, Ikonomovic MD, Styren SD, et al. Upregulation of choline acetyltransferase activity in hippocampus and frontal cortex of elderly subjects with mild cognitive impairment. Ann Neurol. 2002;51:145–55.PubMedCrossRef DeKosky ST, Ikonomovic MD, Styren SD, et al. Upregulation of choline acetyltransferase activity in hippocampus and frontal cortex of elderly subjects with mild cognitive impairment. Ann Neurol. 2002;51:145–55.PubMedCrossRef
19.
go back to reference Mufson EJ, Ma SY, Dills J, et al. Loss of basal forebrain P75(NTR) immunoreactivity in subjects with mild cognitive impairment and Alzheimer’s disease. J Comp Neurol. 2002;443:136–53.PubMedCrossRef Mufson EJ, Ma SY, Dills J, et al. Loss of basal forebrain P75(NTR) immunoreactivity in subjects with mild cognitive impairment and Alzheimer’s disease. J Comp Neurol. 2002;443:136–53.PubMedCrossRef
20.
go back to reference Tiraboschi P, Hansen LA, Alford M, et al. The decline in synapses and cholinergic activity is asynchronous in Alzheimer’s disease. Neurology. 2000;55:1278–83.PubMedCrossRef Tiraboschi P, Hansen LA, Alford M, et al. The decline in synapses and cholinergic activity is asynchronous in Alzheimer’s disease. Neurology. 2000;55:1278–83.PubMedCrossRef
21.
go back to reference Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6:165–77.PubMedCrossRef Geula C, Mesulam MM. Systematic regional variations in the loss of cortical cholinergic fibers in Alzheimer’s disease. Cereb Cortex. 1996;6:165–77.PubMedCrossRef
22.
go back to reference Geula C, Mesulam MM. Cortical cholinergic fibers in aging and Alzheimer’s disease: a morphometric study. Neuroscience. 1989;33:469–81.PubMedCrossRef Geula C, Mesulam MM. Cortical cholinergic fibers in aging and Alzheimer’s disease: a morphometric study. Neuroscience. 1989;33:469–81.PubMedCrossRef
23.
go back to reference Schliebs R, Arendt T. The cholinergic system in aging and neuronal degeneration. Behav Brain Res. 2011;221:555–63.PubMedCrossRef Schliebs R, Arendt T. The cholinergic system in aging and neuronal degeneration. Behav Brain Res. 2011;221:555–63.PubMedCrossRef
25.
go back to reference Candy JM, Perry RH, Perry EK, et al. Pathological changes in the nucleus of Meynert in Alzheimer’s and Parkinson’s diseases. J Neurol Sci. 1983;59:277–89.PubMedCrossRef Candy JM, Perry RH, Perry EK, et al. Pathological changes in the nucleus of Meynert in Alzheimer’s and Parkinson’s diseases. J Neurol Sci. 1983;59:277–89.PubMedCrossRef
26.
go back to reference Nakano I, Hirano A. Parkinson’s disease: neuron loss in the nucleus basalis without concomitant Alzheimer’s disease. Ann Neurol. 1984;15:415–8.PubMedCrossRef Nakano I, Hirano A. Parkinson’s disease: neuron loss in the nucleus basalis without concomitant Alzheimer’s disease. Ann Neurol. 1984;15:415–8.PubMedCrossRef
27.
go back to reference Rogers JD, Brogan D, Mirra SS. The nucleus basalis of Meynert in neurological disease: a quantitative morphological study. Ann Neurol. 1985;17:163–70.PubMedCrossRef Rogers JD, Brogan D, Mirra SS. The nucleus basalis of Meynert in neurological disease: a quantitative morphological study. Ann Neurol. 1985;17:163–70.PubMedCrossRef
28.
go back to reference Tagliavini F, Pilleri G, Bouras C, Constantinidis J. The basal nucleus of Meynert in idiopathic Parkinson’s disease. Acta Neurol Scand. 1984;70:20–8.PubMedCrossRef Tagliavini F, Pilleri G, Bouras C, Constantinidis J. The basal nucleus of Meynert in idiopathic Parkinson’s disease. Acta Neurol Scand. 1984;70:20–8.PubMedCrossRef
29.
go back to reference Whitehouse PJ, Hedreen JC, White III CL, Price DL. Basal forebrain neurons in the dementia of Parkinson disease. Ann Neurol. 1983;13:243–8.PubMedCrossRef Whitehouse PJ, Hedreen JC, White III CL, Price DL. Basal forebrain neurons in the dementia of Parkinson disease. Ann Neurol. 1983;13:243–8.PubMedCrossRef
30.
go back to reference Arendt T, Bigl V, Arendt A, Tennstedt A. Loss of neurons in the nucleus basalis of Meynert in Alzheimer’s disease, paralysis agitans and Korsakoff’s Disease. Acta Neuropathol. 1983;61:101–8.PubMedCrossRef Arendt T, Bigl V, Arendt A, Tennstedt A. Loss of neurons in the nucleus basalis of Meynert in Alzheimer’s disease, paralysis agitans and Korsakoff’s Disease. Acta Neuropathol. 1983;61:101–8.PubMedCrossRef
31.
go back to reference Jellinger K. The pedunculopontine nucleus in Parkinson’s disease, progressive supranuclear palsy and Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1988;51:540–3.PubMedCrossRef Jellinger K. The pedunculopontine nucleus in Parkinson’s disease, progressive supranuclear palsy and Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1988;51:540–3.PubMedCrossRef
32.
go back to reference Gai WP, Halliday GM, Blumbergs PC, et al. Substance P-containing neurons in the mesopontine tegmentum are severely affected in Parkinson’s disease. Brain. 1991;114:2253–67.PubMedCrossRef Gai WP, Halliday GM, Blumbergs PC, et al. Substance P-containing neurons in the mesopontine tegmentum are severely affected in Parkinson’s disease. Brain. 1991;114:2253–67.PubMedCrossRef
33.
go back to reference Hirsch EC, Graybiel AM, Duyckaerts C, Javoy-Agid F. Neuronal loss in the pedunculopontine tegmental nucleus in Parkinson disease and in progressive supranuclear palsy. Proc Natl Acad Sci U S A. 1987;84:5976–80.PubMedCrossRef Hirsch EC, Graybiel AM, Duyckaerts C, Javoy-Agid F. Neuronal loss in the pedunculopontine tegmental nucleus in Parkinson disease and in progressive supranuclear palsy. Proc Natl Acad Sci U S A. 1987;84:5976–80.PubMedCrossRef
34.
go back to reference Zweig RM, Jankel WR, Hedreen JC, et al. The pedunculopontine nucleus in Parkinson’s disease. Ann Neurol. 1989;26:41–6.PubMedCrossRef Zweig RM, Jankel WR, Hedreen JC, et al. The pedunculopontine nucleus in Parkinson’s disease. Ann Neurol. 1989;26:41–6.PubMedCrossRef
35.
go back to reference McKeith IG, Dickson DW, Lowe J, 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, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65:1863–72.PubMedCrossRef
36.
go back to reference Zaccai J, Brayne C, McKeith I, et al. Patterns and stages of alpha-synucleinopathy: relevance in a population-based cohort. Neurology. 2008;70:1042–8.PubMedCrossRef Zaccai J, Brayne C, McKeith I, et al. Patterns and stages of alpha-synucleinopathy: relevance in a population-based cohort. Neurology. 2008;70:1042–8.PubMedCrossRef
37.
go back to reference Perry EK, Irving D, Kerwin JM, et al. Cholinergic transmitter and neurotrophic activities in Lewy body dementia: similarity to Parkinson’s and distinction from Alzheimer disease. Alzheimer Dis Assoc Disord. 1993;7:69–79.PubMedCrossRef Perry EK, Irving D, Kerwin JM, et al. Cholinergic transmitter and neurotrophic activities in Lewy body dementia: similarity to Parkinson’s and distinction from Alzheimer disease. Alzheimer Dis Assoc Disord. 1993;7:69–79.PubMedCrossRef
38.
go back to reference Samuel W, Alford M, Hofstetter CR, Hansen L. Dementia with Lewy bodies vs pure Alzheimer disease: differences in cognition, neuropathology, cholinergic dysfunction, and synapse density. J Neuropathol Exp Neurol. 1997;56:499–508.PubMedCrossRef Samuel W, Alford M, Hofstetter CR, Hansen L. Dementia with Lewy bodies vs pure Alzheimer disease: differences in cognition, neuropathology, cholinergic dysfunction, and synapse density. J Neuropathol Exp Neurol. 1997;56:499–508.PubMedCrossRef
39.
go back to reference Tiraboschi P, Hansen LA, Alford M, et al. Early and widespread cholinergic losses differentiate dementia with Lewy bodies from Alzheimer disease. Arch Gen Psychiatry. 2002;59:946–51.PubMedCrossRef Tiraboschi P, Hansen LA, Alford M, et al. Early and widespread cholinergic losses differentiate dementia with Lewy bodies from Alzheimer disease. Arch Gen Psychiatry. 2002;59:946–51.PubMedCrossRef
40.
go back to reference Tiraboschi P, Hansen LA, Alford M, et al. Cholinergic dysfunction in diseases with Lewy bodies. Neurology. 2000;54:407–11.PubMedCrossRef Tiraboschi P, Hansen LA, Alford M, et al. Cholinergic dysfunction in diseases with Lewy bodies. Neurology. 2000;54:407–11.PubMedCrossRef
41.
go back to reference Wenning GK, Ebersbach G, Verny M, et al. Progression of falls in postmortem-confirmed parkinsonian disorders. Mov Disord. 1999;14:947–50.PubMedCrossRef Wenning GK, Ebersbach G, Verny M, et al. Progression of falls in postmortem-confirmed parkinsonian disorders. Mov Disord. 1999;14:947–50.PubMedCrossRef
42.
go back to reference Shinotoh H, Namba H, Yamaguchi M, et al. Positron emission tomographic measurement of acetylcholinesterase activity reveals differential loss of ascending cholinergic systems in Parkinson’s disease and progressive supranuclear palsy. Ann Neurol. 1999;46:62–9.PubMedCrossRef Shinotoh H, Namba H, Yamaguchi M, et al. Positron emission tomographic measurement of acetylcholinesterase activity reveals differential loss of ascending cholinergic systems in Parkinson’s disease and progressive supranuclear palsy. Ann Neurol. 1999;46:62–9.PubMedCrossRef
43.
go back to reference Gilman S, Koeppe RA, Nan B, et al. Cerebral cortical and subcortical cholinergic deficits in parkinsonian syndromes. Neurology. 2010;74:1416–23.PubMedCrossRef Gilman S, Koeppe RA, Nan B, et al. Cerebral cortical and subcortical cholinergic deficits in parkinsonian syndromes. Neurology. 2010;74:1416–23.PubMedCrossRef
44.
go back to reference Mesulam MM, Geula C. Overlap between acetylcholinesterase-rich and choline acetyltransferase-positive (cholinergic) axons in human cerebral cortex. Brain Res. 1992;577:112–20.PubMedCrossRef Mesulam MM, Geula C. Overlap between acetylcholinesterase-rich and choline acetyltransferase-positive (cholinergic) axons in human cerebral cortex. Brain Res. 1992;577:112–20.PubMedCrossRef
45.
go back to reference Atack JR, Perry EK, Bonham JR, et al. Molecular forms of acetylcholinesterase and butyrylcholinesterase in the aged human central nervous system. J Neurochem. 1986;47:263–77.PubMedCrossRef Atack JR, Perry EK, Bonham JR, et al. Molecular forms of acetylcholinesterase and butyrylcholinesterase in the aged human central nervous system. J Neurochem. 1986;47:263–77.PubMedCrossRef
46.
go back to reference Hilker R, Thomas AV, Klein JC, et al. Dementia in Parkinson disease: functional imaging of cholinergic and dopaminergic pathways. Neurology. 2005;65:1716–22.PubMedCrossRef Hilker R, Thomas AV, Klein JC, et al. Dementia in Parkinson disease: functional imaging of cholinergic and dopaminergic pathways. Neurology. 2005;65:1716–22.PubMedCrossRef
47.
go back to reference Kuhl DE, Minoshima S, Fessler JA, et al. In vivo mapping of cholinergic terminals in normal aging, Alzheimer’s disease, and Parkinson’s disease. Ann Neurol. 1996;40:399–410.PubMedCrossRef Kuhl DE, Minoshima S, Fessler JA, et al. In vivo mapping of cholinergic terminals in normal aging, Alzheimer’s disease, and Parkinson’s disease. Ann Neurol. 1996;40:399–410.PubMedCrossRef
48.
go back to reference Shimada H, Hirano S, Shinotoh H, et al. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET. Neurology. 2009;73:273–8.PubMedCrossRef Shimada H, Hirano S, Shinotoh H, et al. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET. Neurology. 2009;73:273–8.PubMedCrossRef
49.
go back to reference Klein JC, Eggers C, Kalbe E, et al. Neurotransmitter changes in dementia with Lewy bodies and Parkinson disease dementia in vivo. Neurology. 2010;74:885–92.PubMedCrossRef Klein JC, Eggers C, Kalbe E, et al. Neurotransmitter changes in dementia with Lewy bodies and Parkinson disease dementia in vivo. Neurology. 2010;74:885–92.PubMedCrossRef
50.
go back to reference Marcone A, Garibotto V, Moresco RM, et al. [11C]-MP4A PET cholinergic measurements in amnestic mild cognitive impairment, probable Alzheimer’s disease, and dementia with Lewy bodies: a Bayesian method and voxel-based analysis. J Alzheimers Dis. 2012;31:387–99.PubMed Marcone A, Garibotto V, Moresco RM, et al. [11C]-MP4A PET cholinergic measurements in amnestic mild cognitive impairment, probable Alzheimer’s disease, and dementia with Lewy bodies: a Bayesian method and voxel-based analysis. J Alzheimers Dis. 2012;31:387–99.PubMed
51.
go back to reference Pimlott SL, Piggott M, Owens J, et al. Nicotinic acetylcholine receptor distribution in Alzheimer’s disease, dementia with Lewy bodies, Parkinson’s disease, and vascular dementia: in vitro binding study using 5-[125I]-A-85380. Neuropsychopharmacology. 2004;29:108–16.PubMedCrossRef Pimlott SL, Piggott M, Owens J, et al. Nicotinic acetylcholine receptor distribution in Alzheimer’s disease, dementia with Lewy bodies, Parkinson’s disease, and vascular dementia: in vitro binding study using 5-[125I]-A-85380. Neuropsychopharmacology. 2004;29:108–16.PubMedCrossRef
52.
go back to reference Kas A, Bottlaender M, Gallezot JD, et al. Decrease of nicotinic receptors in the nigrostriatal system in Parkinson’s disease. J Cereb Blood Flow Metab. 2009;29:1601–8.PubMedCrossRef Kas A, Bottlaender M, Gallezot JD, et al. Decrease of nicotinic receptors in the nigrostriatal system in Parkinson’s disease. J Cereb Blood Flow Metab. 2009;29:1601–8.PubMedCrossRef
53.
go back to reference Fujita M, Ichise M, Zoghbi SS, et al. Widespread decrease of nicotinic acetylcholine receptors in Parkinson’s disease. Ann Neurol. 2006;59:174–7.PubMedCrossRef Fujita M, Ichise M, Zoghbi SS, et al. Widespread decrease of nicotinic acetylcholine receptors in Parkinson’s disease. Ann Neurol. 2006;59:174–7.PubMedCrossRef
54.
go back to reference Meyer PM, Strecker K, Kendziorra K, et al. Reduced α4β2*-nicotinic acetylcholine receptor binding and its relationship to mild cognitive and depressive symptoms in Parkinson disease. Arch Gen Psychiatry. 2009;66:866–77.PubMedCrossRef Meyer PM, Strecker K, Kendziorra K, et al. Reduced α4β2*-nicotinic acetylcholine receptor binding and its relationship to mild cognitive and depressive symptoms in Parkinson disease. Arch Gen Psychiatry. 2009;66:866–77.PubMedCrossRef
55.
go back to reference Asahina M, Suhara T, Shinotoh H, et al. Brain muscarinic receptors in progressive supranuclear palsy and Parkinson’s disease: a positron emission tomographic study. J Neurol Neurosurg Psychiatry. 1998;65:155–63.PubMedCrossRef Asahina M, Suhara T, Shinotoh H, et al. Brain muscarinic receptors in progressive supranuclear palsy and Parkinson’s disease: a positron emission tomographic study. J Neurol Neurosurg Psychiatry. 1998;65:155–63.PubMedCrossRef
56.
go back to reference • Bohnen NI, Muller ML, Kotagal V, et al. Heterogeneity of cholinergic denervation in Parkinson’s disease without dementia. J Cereb Blood Flow Metab. 2012;32:1609–17. This study shows that cholinergic loss is variable in nondemented PD patients. The heterogeneity of cholinergic losses across nondemented patients suggests that cholinergic treatment in these patients should be targeted.PubMedCrossRef • Bohnen NI, Muller ML, Kotagal V, et al. Heterogeneity of cholinergic denervation in Parkinson’s disease without dementia. J Cereb Blood Flow Metab. 2012;32:1609–17. This study shows that cholinergic loss is variable in nondemented PD patients. The heterogeneity of cholinergic losses across nondemented patients suggests that cholinergic treatment in these patients should be targeted.PubMedCrossRef
57.
go back to reference Aarsland D, Kurz MW. The epidemiology of dementia associated with Parkinson disease. J Neurol Sci. 2010;289:18–22.PubMedCrossRef Aarsland D, Kurz MW. The epidemiology of dementia associated with Parkinson disease. J Neurol Sci. 2010;289:18–22.PubMedCrossRef
58.
go back to reference Koerts J, Leenders KL, Brouwer WH. Cognitive dysfunction in nondemented Parkinson’s disease patients: controlled and automatic behavior. Cortex. 2009;45:922–9.PubMedCrossRef Koerts J, Leenders KL, Brouwer WH. Cognitive dysfunction in nondemented Parkinson’s disease patients: controlled and automatic behavior. Cortex. 2009;45:922–9.PubMedCrossRef
59.
go back to reference Sawamoto N, Piccini P, Hotton G, et al. Cognitive deficits and striato-frontal dopamine release in Parkinson’s disease. Brain. 2008;131:1294–302.PubMedCrossRef Sawamoto N, Piccini P, Hotton G, et al. Cognitive deficits and striato-frontal dopamine release in Parkinson’s disease. Brain. 2008;131:1294–302.PubMedCrossRef
60.
go back to reference Cools R. Dopaminergic modulation of cognitive function-implications for L-DOPA treatment in Parkinson’s disease. Neurosci Biobehav Rev. 2006;30:1–23.PubMedCrossRef Cools R. Dopaminergic modulation of cognitive function-implications for L-DOPA treatment in Parkinson’s disease. Neurosci Biobehav Rev. 2006;30:1–23.PubMedCrossRef
61.
go back to reference Ruberg M, Rieger F, Villageois A, et al. Acetylcholinesterase and butyrylcholinesterase in frontal cortex and cerebrospinal fluid of demented and nondemented patients with Parkinson’s disease. Brain Res. 1986;362:83–91.PubMedCrossRef Ruberg M, Rieger F, Villageois A, et al. Acetylcholinesterase and butyrylcholinesterase in frontal cortex and cerebrospinal fluid of demented and nondemented patients with Parkinson’s disease. Brain Res. 1986;362:83–91.PubMedCrossRef
62.
go back to reference Mattila PM, Roytta M, Lonnberg P, et al. Choline acetytransferase activity and striatal dopamine receptors in Parkinson’s disease in relation to cognitive impairment. Acta Neuropathol. 2001;102:160–6.PubMed Mattila PM, Roytta M, Lonnberg P, et al. Choline acetytransferase activity and striatal dopamine receptors in Parkinson’s disease in relation to cognitive impairment. Acta Neuropathol. 2001;102:160–6.PubMed
63.
go back to reference Bohnen NI, Kaufer DI, Hendrickson R, et al. Cognitive correlates of cortical cholinergic denervation in Parkinson’s disease and parkinsonian dementia. J Neurol. 2006;253:242–7.PubMedCrossRef Bohnen NI, Kaufer DI, Hendrickson R, et al. Cognitive correlates of cortical cholinergic denervation in Parkinson’s disease and parkinsonian dementia. J Neurol. 2006;253:242–7.PubMedCrossRef
64.
go back to reference Kehagia AA, Barker RA, Robbins TW. Cognitive impairment in Parkinson’s disease: the dual syndrome hypothesis. Neurodegener Dis. 2013;11:79–92.PubMedCrossRef Kehagia AA, Barker RA, Robbins TW. Cognitive impairment in Parkinson’s disease: the dual syndrome hypothesis. Neurodegener Dis. 2013;11:79–92.PubMedCrossRef
65.
go back to reference Williams-Gray CH, Evans JR, Goris A, et al. The distinct cognitive syndromes of Parkinson’s disease: 5-year follow-up of the CamPaIGN cohort. Brain. 2009;132:2958–69.PubMedCrossRef Williams-Gray CH, Evans JR, Goris A, et al. The distinct cognitive syndromes of Parkinson’s disease: 5-year follow-up of the CamPaIGN cohort. Brain. 2009;132:2958–69.PubMedCrossRef
66.
go back to reference Aubert I, Araujo DM, Cecyre D, et al. Comparative alterations of nicotinic and muscarinic binding sites in Alzheimer’s and Parkinson’s diseases. J Neurochem. 1992;58:529–41.PubMedCrossRef Aubert I, Araujo DM, Cecyre D, et al. Comparative alterations of nicotinic and muscarinic binding sites in Alzheimer’s and Parkinson’s diseases. J Neurochem. 1992;58:529–41.PubMedCrossRef
67.
go back to reference Whitehouse PJ, Martino AM, Wagster MV, et al. Reductions in [3H]nicotinic acetylcholine binding in Alzheimer’s disease and Parkinson’s disease: an autoradiographic study. Neurology. 1988;38:720–3.PubMedCrossRef Whitehouse PJ, Martino AM, Wagster MV, et al. Reductions in [3H]nicotinic acetylcholine binding in Alzheimer’s disease and Parkinson’s disease: an autoradiographic study. Neurology. 1988;38:720–3.PubMedCrossRef
68.
go back to reference Petrou M, Kotagal V, Bohnen NI. An update on brain imaging in parkinsonian dementia. Imaging Med. 2012;4:201–13.PubMedCrossRef Petrou M, Kotagal V, Bohnen NI. An update on brain imaging in parkinsonian dementia. Imaging Med. 2012;4:201–13.PubMedCrossRef
69.
go back to reference Kulisevsky J, Pagonabarraga J, Pascual-Sedano B, et al. Prevalence and correlates of neuropsychiatric symptoms in Parkinson’s disease without dementia. Mov Disord. 2008;23:1889–96.PubMedCrossRef Kulisevsky J, Pagonabarraga J, Pascual-Sedano B, et al. Prevalence and correlates of neuropsychiatric symptoms in Parkinson’s disease without dementia. Mov Disord. 2008;23:1889–96.PubMedCrossRef
70.
go back to reference Bohnen NI, Kaufer DI, Hendrickson R, et al. Cortical cholinergic denervation is associated with depressive symptoms in Parkinson’s disease and parkinsonian dementia. J Neurol Neurosurg Psychiatry. 2007;78:641–3.PubMedCrossRef Bohnen NI, Kaufer DI, Hendrickson R, et al. Cortical cholinergic denervation is associated with depressive symptoms in Parkinson’s disease and parkinsonian dementia. J Neurol Neurosurg Psychiatry. 2007;78:641–3.PubMedCrossRef
71.
go back to reference Troster AI, Stalp LD, Paolo AM, et al. Neuropsychological impairment in Parkinson’s disease with and without depression. Arch Neurol. 1995;52:1164–9.PubMedCrossRef Troster AI, Stalp LD, Paolo AM, et al. Neuropsychological impairment in Parkinson’s disease with and without depression. Arch Neurol. 1995;52:1164–9.PubMedCrossRef
72.
go back to reference Mayeux R, Stern Y, Rosen J, Leventhal J. Depression, intellectual impairment, and Parkinson disease. Neurology. 1981;31:645–50.PubMedCrossRef Mayeux R, Stern Y, Rosen J, Leventhal J. Depression, intellectual impairment, and Parkinson disease. Neurology. 1981;31:645–50.PubMedCrossRef
73.
go back to reference Lieberman A. Are dementia and depression in Parkinson’s disease related? J Neurol Sci. 2006;248:138–42.PubMedCrossRef Lieberman A. Are dementia and depression in Parkinson’s disease related? J Neurol Sci. 2006;248:138–42.PubMedCrossRef
74.
go back to reference Weintraub D, Morales KH, Moberg PJ, et al. Antidepressant studies in Parkinson’s disease: a review and meta-analysis. Mov Disord. 2005;20:1161–9.PubMedCrossRef Weintraub D, Morales KH, Moberg PJ, et al. Antidepressant studies in Parkinson’s disease: a review and meta-analysis. Mov Disord. 2005;20:1161–9.PubMedCrossRef
75.
go back to reference Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease—a multicenter study. Parkinsonism Relat Disord. 2009;15:490–4.PubMedCrossRef Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease—a multicenter study. Parkinsonism Relat Disord. 2009;15:490–4.PubMedCrossRef
77.
go back to reference Bohnen NI, Muller ML. In vivo neurochemical imaging of olfactory dysfunction in Parkinson’s disease. J Neural Transm. 2013;120:571–6.PubMedCrossRef Bohnen NI, Muller ML. In vivo neurochemical imaging of olfactory dysfunction in Parkinson’s disease. J Neural Transm. 2013;120:571–6.PubMedCrossRef
78.
go back to reference Bohnen NI, Muller ML, Kotagal V, et al. Olfactory dysfunction, central cholinergic integrity and cognitive impairment in Parkinson’s disease. Brain. 2010;133:1747–54.PubMedCrossRef Bohnen NI, Muller ML, Kotagal V, et al. Olfactory dysfunction, central cholinergic integrity and cognitive impairment in Parkinson’s disease. Brain. 2010;133:1747–54.PubMedCrossRef
79.
go back to reference Bohnen NI, Gedela S, Herath P, et al. Selective hyposmia in Parkinson disease: association with hippocampal dopamine activity. Neurosci Lett. 2008;447:12–6.PubMedCrossRef Bohnen NI, Gedela S, Herath P, et al. Selective hyposmia in Parkinson disease: association with hippocampal dopamine activity. Neurosci Lett. 2008;447:12–6.PubMedCrossRef
80.
go back to reference Marion MH, Qurashi M, Marshall G, Foster O. Is REM sleep behaviour disorder (RBD) a risk factor of dementia in idiopathic Parkinson’s disease? J Neurol. 2008;255:192–6.PubMedCrossRef Marion MH, Qurashi M, Marshall G, Foster O. Is REM sleep behaviour disorder (RBD) a risk factor of dementia in idiopathic Parkinson’s disease? J Neurol. 2008;255:192–6.PubMedCrossRef
81.
go back to reference Vendette M, Gagnon JF, Decary A, et al. REM sleep behavior disorder predicts cognitive impairment in Parkinson disease without dementia. Neurology. 2007;69:1843–9.PubMedCrossRef Vendette M, Gagnon JF, Decary A, et al. REM sleep behavior disorder predicts cognitive impairment in Parkinson disease without dementia. Neurology. 2007;69:1843–9.PubMedCrossRef
82.
go back to reference Postuma RB, Gagnon JF, Montplaisir J. Cognition in REM sleep behavior disorder—a window into preclinical dementia? Sleep Med. 2008;9:341–2.PubMedCrossRef Postuma RB, Gagnon JF, Montplaisir J. Cognition in REM sleep behavior disorder—a window into preclinical dementia? Sleep Med. 2008;9:341–2.PubMedCrossRef
83.
go back to reference • Kotagal V, Albin RL, Muller ML, et al. Symptoms of rapid eye movement sleep behavior disorder are associated with cholinergic denervation in Parkinson disease. Ann Neurol. 2012;71:560–8. To the best of our knowledge this is the first in vivo PET study to show that REM sleep behavior disorder (RBD) is associated with cholinergic denervation in PD. RBD is one of the first prodromal symptoms of Lewy body disorders. This result may indicate early cholinergic system degeneration in some patients.PubMedCrossRef • Kotagal V, Albin RL, Muller ML, et al. Symptoms of rapid eye movement sleep behavior disorder are associated with cholinergic denervation in Parkinson disease. Ann Neurol. 2012;71:560–8. To the best of our knowledge this is the first in vivo PET study to show that REM sleep behavior disorder (RBD) is associated with cholinergic denervation in PD. RBD is one of the first prodromal symptoms of Lewy body disorders. This result may indicate early cholinergic system degeneration in some patients.PubMedCrossRef
84.
go back to reference Postuma RB, Gagnon JF, Montplaisir JY. REM sleep behavior disorder: from dreams to neurodegeneration. Neurobiol Dis. 2012;46:553–8.PubMedCrossRef Postuma RB, Gagnon JF, Montplaisir JY. REM sleep behavior disorder: from dreams to neurodegeneration. Neurobiol Dis. 2012;46:553–8.PubMedCrossRef
85.
go back to reference Muslimovic D, Post B, Speelman JD, et al. Determinants of disability and quality of life in mild to moderate Parkinson disease. Neurology. 2008;70:2241–7.PubMedCrossRef Muslimovic D, Post B, Speelman JD, et al. Determinants of disability and quality of life in mild to moderate Parkinson disease. Neurology. 2008;70:2241–7.PubMedCrossRef
86.
go back to reference Sethi K. Levodopa unresponsive symptoms in Parkinson disease. Mov Disord. 2008;23 Suppl 3:S521–33.PubMedCrossRef Sethi K. Levodopa unresponsive symptoms in Parkinson disease. Mov Disord. 2008;23 Suppl 3:S521–33.PubMedCrossRef
87.
go back to reference •• Karachi C, Grabli D, Bernard FA, et al. Cholinergic mesencephalic neurons are involved in gait and postural disorders in Parkinson disease. J Clin Invest. 2010;120:2745–54. An elegant study that convincingly shows the role of the PPN in mobility impairment in parkinsonian monkeys and human patients with PD. Evidence for this is described in 3-fold in complementary human fMRI, human post-mortem, and primate lesioning studies. The finding that posture and gait deficits can occur with isolated PPN impairment in the absence of nigrostriatal dopaminergic lesions has major implications for current understanding of mobility impairments in PD.PubMedCrossRef •• Karachi C, Grabli D, Bernard FA, et al. Cholinergic mesencephalic neurons are involved in gait and postural disorders in Parkinson disease. J Clin Invest. 2010;120:2745–54. An elegant study that convincingly shows the role of the PPN in mobility impairment in parkinsonian monkeys and human patients with PD. Evidence for this is described in 3-fold in complementary human fMRI, human post-mortem, and primate lesioning studies. The finding that posture and gait deficits can occur with isolated PPN impairment in the absence of nigrostriatal dopaminergic lesions has major implications for current understanding of mobility impairments in PD.PubMedCrossRef
88.
go back to reference Bohnen NI, Muller ML, Koeppe RA, et al. History of falls in Parkinson disease is associated with reduced cholinergic activity. Neurology. 2009;73:1670–6.PubMedCrossRef Bohnen NI, Muller ML, Koeppe RA, et al. History of falls in Parkinson disease is associated with reduced cholinergic activity. Neurology. 2009;73:1670–6.PubMedCrossRef
89.
go back to reference Bakker M, de Lange FP, Stevens JA, et al. Motor imagery of gait: a quantitative approach. Exp Brain Res. 2007;179:497–504.PubMedCrossRef Bakker M, de Lange FP, Stevens JA, et al. Motor imagery of gait: a quantitative approach. Exp Brain Res. 2007;179:497–504.PubMedCrossRef
90.
go back to reference Cremers J, D’Ostilio K, Stamatakis J, et al. Brain activation pattern related to gait disturbances in Parkinson’s disease. Mov Disord. 2012;27:1498–505.PubMedCrossRef Cremers J, D’Ostilio K, Stamatakis J, et al. Brain activation pattern related to gait disturbances in Parkinson’s disease. Mov Disord. 2012;27:1498–505.PubMedCrossRef
91.
go back to reference Snijders AH, Leunissen I, Bakker M, et al. Gait-related cerebral alterations in patients with Parkinson’s disease with freezing of gait. Brain. 2011;134:59–72.PubMedCrossRef Snijders AH, Leunissen I, Bakker M, et al. Gait-related cerebral alterations in patients with Parkinson’s disease with freezing of gait. Brain. 2011;134:59–72.PubMedCrossRef
92.
go back to reference Bohnen N, Kotagal V, Albin R, et al. Gait speed is preserved in oligosystem compared with multisystem neurodegeneration in Parkinson disease. Neurology. 2013;80, P04165. Bohnen N, Kotagal V, Albin R, et al. Gait speed is preserved in oligosystem compared with multisystem neurodegeneration in Parkinson disease. Neurology. 2013;80, P04165.
93.
go back to reference Woollacott M, Shumway-Cook A. Attention and the control of posture and gait: a review of an emerging area of research. Gait Posture. 2002;16:1–14.PubMedCrossRef Woollacott M, Shumway-Cook A. Attention and the control of posture and gait: a review of an emerging area of research. Gait Posture. 2002;16:1–14.PubMedCrossRef
94.
go back to reference Yarnall AJ, Rochester L, Baker MR, et al. Short latency afferent inhibition: a biomarker for mild cognitive impairment in Parkinson’s disease? Mov Disord. 2013. doi:10.1002/mds.25360. Yarnall AJ, Rochester L, Baker MR, et al. Short latency afferent inhibition: a biomarker for mild cognitive impairment in Parkinson’s disease? Mov Disord. 2013. doi:10.​1002/​mds.​25360.
95.
go back to reference Rochester L, Yarnall AJ, Baker MR, et al. Cholinergic dysfunction contributes to gait disturbance in early Parkinson’s disease. Brain. 2012;135:2779–88.PubMedCrossRef Rochester L, Yarnall AJ, Baker MR, et al. Cholinergic dysfunction contributes to gait disturbance in early Parkinson’s disease. Brain. 2012;135:2779–88.PubMedCrossRef
96.
go back to reference Albin RL, Koeppe RA, Bohnen NI, et al. Spared caudal brainstem SERT binding in early Parkinson’s disease. J Cereb Blood Flow Metab. 2008;28:441–4.PubMedCrossRef Albin RL, Koeppe RA, Bohnen NI, et al. Spared caudal brainstem SERT binding in early Parkinson’s disease. J Cereb Blood Flow Metab. 2008;28:441–4.PubMedCrossRef
97.
go back to reference Maetzler W, Liepelt I, Reimold M, et al. Cortical PIB binding in Lewy body disease is associated with Alzheimer-like characteristics. Neurobiol Dis. 2009;34:107–12.PubMedCrossRef Maetzler W, Liepelt I, Reimold M, et al. Cortical PIB binding in Lewy body disease is associated with Alzheimer-like characteristics. Neurobiol Dis. 2009;34:107–12.PubMedCrossRef
98.
go back to reference Maetzler W, Reimold M, Liepelt I, et al. [11C]PIB binding in Parkinson’s disease dementia. NeuroImage. 2008;39:1027–33.PubMedCrossRef Maetzler W, Reimold M, Liepelt I, et al. [11C]PIB binding in Parkinson’s disease dementia. NeuroImage. 2008;39:1027–33.PubMedCrossRef
99.
go back to reference Mann DM, Yates PO, Hawkes J. The pathology of the human locus ceruleus. Clin Neuropathol. 1983;2:1–7.PubMed Mann DM, Yates PO, Hawkes J. The pathology of the human locus ceruleus. Clin Neuropathol. 1983;2:1–7.PubMed
100.
go back to reference Muller ML, Frey KA, Petrou M, et al. β-amyloid and postural instability and gait difficulty in Parkinson’s disease at risk for dementia. Mov Disord. 2013;28:296–301.PubMedCrossRef Muller ML, Frey KA, Petrou M, et al. β-amyloid and postural instability and gait difficulty in Parkinson’s disease at risk for dementia. Mov Disord. 2013;28:296–301.PubMedCrossRef
Metadata
Title
Cholinergic Dysfunction in Parkinson’s Disease
Authors
Martijn L. T. M. Müller
Nicolaas I. Bohnen
Publication date
01-09-2013
Publisher
Springer US
Published in
Current Neurology and Neuroscience Reports / Issue 9/2013
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-013-0377-9

Other articles of this Issue 9/2013

Current Neurology and Neuroscience Reports 9/2013 Go to the issue