Skip to main content
Erschienen in: Current Allergy and Asthma Reports 8/2018

01.08.2018 | Rhinosinusitis (J Mullol, Section Editor)

Olfactory Dysfunction in Neurodegenerative Diseases

verfasst von: Concepció Marin, Dolores Vilas, Cristóbal Langdon, Isam Alobid, Mauricio López-Chacón, Antje Haehner, Thomas Hummel, Joaquim Mullol

Erschienen in: Current Allergy and Asthma Reports | Ausgabe 8/2018

Einloggen, um Zugang zu erhalten

Abstract

Purpose of Review

The sense of smell is today one of the focuses of interest in aging and neurodegenerative disease research. In several neurodegenerative diseases, such as Parkinson’s disease and Alzheimer’s disease, the olfactory dysfunction is one of the initial symptoms appearing years before motor symptoms and cognitive decline, being considered a clinical marker of these diseases’ early stages and a marker of disease progression and cognitive decline. Overall and under the umbrella of precision medicine, attention to olfactory function may help to improve chances of success for neuroprotective and disease-modifying therapeutic strategies.

Recent Findings

The use of olfaction, as clinical marker for neurodegenerative diseases is helpful in the characterization of prodromal stages of these diseases, early diagnostic strategies, differential diagnosis, and potentially prediction of treatment success. Understanding the mechanisms underlying olfactory dysfunction is central to determine its association with neurodegenerative disorders. Several anatomical systems and environmental factors may underlie or contribute to olfactory loss associated with neurological diseases, although the direct biological link to each disorder remains unclear and, thus, requires further investigation.

Summary

In this review, we describe the neurobiology of olfaction, and the most common olfactory function measurements in neurodegenerative diseases. We also highlight the evidence for the presence of olfactory dysfunction in several neurodegenerative diseases, its value as a clinical marker for early stages of the diseases when combined with other clinical, biological, and neuroimage markers, and its role as a useful symptom for the differential diagnosis and follow-up of disease. The neuropathological correlations and the changes in neurotransmitter systems related with olfactory dysfunction in the neurodegenerative diseases are also described.
Literatur
1.
Zurück zum Zitat Hummel T, Landis BN, Hüttenbrik KB. Smell and taste disorders. GMS Curr Top Otorhinolaryngol Head Neck Surg. 2011;10:Doc04.PubMed Hummel T, Landis BN, Hüttenbrik KB. Smell and taste disorders. GMS Curr Top Otorhinolaryngol Head Neck Surg. 2011;10:Doc04.PubMed
2.
Zurück zum Zitat Doty RL, Kamath V. The influences of age on olfaction: a review. Front Psychol. 2014;5:1–20.CrossRef Doty RL, Kamath V. The influences of age on olfaction: a review. Front Psychol. 2014;5:1–20.CrossRef
3.
Zurück zum Zitat Ottaviano G, Zuccarello D, Frasson G, Scarpa B, Nardello E, Foresta C, et al. Olfactory sensitivity and sexual desire in young adult and elderly men: an introductory investigation. Am J Rhinol Allergy. 2013;27:157–61.PubMedCrossRef Ottaviano G, Zuccarello D, Frasson G, Scarpa B, Nardello E, Foresta C, et al. Olfactory sensitivity and sexual desire in young adult and elderly men: an introductory investigation. Am J Rhinol Allergy. 2013;27:157–61.PubMedCrossRef
4.
Zurück zum Zitat Ottaviano G, Frasson G, Nardello E, Martini A. Olfaction deterioration in cognitive disorders in the elderly. Aging Clin Exp Res. 2016;28:37–45.PubMedCrossRef Ottaviano G, Frasson G, Nardello E, Martini A. Olfaction deterioration in cognitive disorders in the elderly. Aging Clin Exp Res. 2016;28:37–45.PubMedCrossRef
5.
6.
Zurück zum Zitat Mullol J, Alobid I, Mariño-Sánchez F, Quintó L, de Haro J, Bernal-Sprekelsen M, et al. Furthering the understanding of olfaction, prevalence of loss of smell and risk factors: a population-based survey (OLFACAT study). BMJ Open. 2012;2:e001256.PubMedPubMedCentralCrossRef Mullol J, Alobid I, Mariño-Sánchez F, Quintó L, de Haro J, Bernal-Sprekelsen M, et al. Furthering the understanding of olfaction, prevalence of loss of smell and risk factors: a population-based survey (OLFACAT study). BMJ Open. 2012;2:e001256.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Rey NL, Wesson DW, Brundin P. The olfactory bulb as the entry site for prion-like propagation in neurodegenerative diseases. Neurobiol Dis. 2018;109:226–48.PubMedCrossRef Rey NL, Wesson DW, Brundin P. The olfactory bulb as the entry site for prion-like propagation in neurodegenerative diseases. Neurobiol Dis. 2018;109:226–48.PubMedCrossRef
9.
Zurück zum Zitat Doty RL. Olfactory dysfunction in neurodegenerative diseases: is there a common pathological substrates? Lancet Neurol. 2017;16:478–88.PubMedCrossRef Doty RL. Olfactory dysfunction in neurodegenerative diseases: is there a common pathological substrates? Lancet Neurol. 2017;16:478–88.PubMedCrossRef
10.
Zurück zum Zitat Mariño-Sánchez FS, Alobid I, Centellas S, Alberca C, Guilemany JM, Canals JM, et al. Smell training increases cognitive smell skills of wine tasters compared to the general healthy population. The WINECAT Study. Rhinology. 2010;48:273–6.PubMedCrossRef Mariño-Sánchez FS, Alobid I, Centellas S, Alberca C, Guilemany JM, Canals JM, et al. Smell training increases cognitive smell skills of wine tasters compared to the general healthy population. The WINECAT Study. Rhinology. 2010;48:273–6.PubMedCrossRef
11.
Zurück zum Zitat Banks SJ, Sreenivasan KR, Weintraub DM, et al. Structural and functional MRI differences in master sommeliers: a pilot study on expertise in the brain. Front Hum Neurosci. 2016;10:414.PubMedPubMedCentralCrossRef Banks SJ, Sreenivasan KR, Weintraub DM, et al. Structural and functional MRI differences in master sommeliers: a pilot study on expertise in the brain. Front Hum Neurosci. 2016;10:414.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Delon-Martin C, Palilly J, Fonlupt P, Veyrac A, Royet JP. Perfurmers’ expertise induces structural reorganization in olfactory brain regions. Neuroimage. 2013;68:55–62.PubMedCrossRef Delon-Martin C, Palilly J, Fonlupt P, Veyrac A, Royet JP. Perfurmers’ expertise induces structural reorganization in olfactory brain regions. Neuroimage. 2013;68:55–62.PubMedCrossRef
13.
Zurück zum Zitat Frasnelli J, Hummel T. Olfactory dysfunction and daily life. Eur Arch Otorhinolaryngol. 2005;262:231–5.PubMedCrossRef Frasnelli J, Hummel T. Olfactory dysfunction and daily life. Eur Arch Otorhinolaryngol. 2005;262:231–5.PubMedCrossRef
14.
Zurück zum Zitat Attems J, Wlaker L, Jellinger KA. Olfaction and aging: a mini-review. Gerontology. 2015;61:485–90.PubMedCrossRef Attems J, Wlaker L, Jellinger KA. Olfaction and aging: a mini-review. Gerontology. 2015;61:485–90.PubMedCrossRef
15.
Zurück zum Zitat Vassilaki M, Christianson TJ, Mielke MM, et al. Neuroimaging biomarkers and impaired olfaction in cognitively normal individuals. Ann Neurol. 2017;81:871–82.PubMedPubMedCentralCrossRef Vassilaki M, Christianson TJ, Mielke MM, et al. Neuroimaging biomarkers and impaired olfaction in cognitively normal individuals. Ann Neurol. 2017;81:871–82.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Temmel AF, Quint C, Schickinger-Fischer B, Klimek L, Stoller E, Hummel T. Characteristics of olfactory disorders in relation to major causes of olfactory loss. Arch Otolaryngol Head Neck Surg. 2002;128:635–41.PubMedCrossRef Temmel AF, Quint C, Schickinger-Fischer B, Klimek L, Stoller E, Hummel T. Characteristics of olfactory disorders in relation to major causes of olfactory loss. Arch Otolaryngol Head Neck Surg. 2002;128:635–41.PubMedCrossRef
17.
Zurück zum Zitat Hoffman HJ, Rawal S, Li CM, Duffy VB. New chemosensory component in the US National Health and Nutrition Examination Survey (NHANES): first-year results for measured olfactory dysfunction. Rev Endrocr Metab Disord. 2016;17:221–40.CrossRef Hoffman HJ, Rawal S, Li CM, Duffy VB. New chemosensory component in the US National Health and Nutrition Examination Survey (NHANES): first-year results for measured olfactory dysfunction. Rev Endrocr Metab Disord. 2016;17:221–40.CrossRef
18.
Zurück zum Zitat Hummel T, Nordin S. Olfatory disorders and their consequences for quality of life. Acta Otolaryngol. 2005;125:116–21.PubMedCrossRef Hummel T, Nordin S. Olfatory disorders and their consequences for quality of life. Acta Otolaryngol. 2005;125:116–21.PubMedCrossRef
19.
Zurück zum Zitat Pence TS, Reiter ER, DiNardo LJ, Costanzo RM. Risk factors for hazardous events in olfactory-impaired patients. JAMA Otolaryngol Head Neck Surg. 2014;140:951–5.PubMedCrossRef Pence TS, Reiter ER, DiNardo LJ, Costanzo RM. Risk factors for hazardous events in olfactory-impaired patients. JAMA Otolaryngol Head Neck Surg. 2014;140:951–5.PubMedCrossRef
20.
Zurück zum Zitat Jaume F, Quintó L, Alobid J, Mullol J. Overuse of diagnostic tools and medications in acute rhinosinusitis in Spain: a population-based study (the PROSINUS study). BMJ Open. 2018;8:e018788.PubMedPubMedCentralCrossRef Jaume F, Quintó L, Alobid J, Mullol J. Overuse of diagnostic tools and medications in acute rhinosinusitis in Spain: a population-based study (the PROSINUS study). BMJ Open. 2018;8:e018788.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Langdon C, Guillemany JM, Valls M, Alobid I, Bartra J, Dávila I, et al. Allergic rhinitis causes loss of smell in children. Pediatr Allergy Immunol. 2016;27:867–70.PubMedCrossRef Langdon C, Guillemany JM, Valls M, Alobid I, Bartra J, Dávila I, et al. Allergic rhinitis causes loss of smell in children. Pediatr Allergy Immunol. 2016;27:867–70.PubMedCrossRef
22.
Zurück zum Zitat Langdon C, Lehrer E, Berenguer J, Laxe S, Alobid I, Quintó L, et al. Olfactory training in posttraumatic smell impairment: mild improvement in threshold performances-results from a randomized controlled study. J Neurotrauma. 2018; Langdon C, Lehrer E, Berenguer J, Laxe S, Alobid I, Quintó L, et al. Olfactory training in posttraumatic smell impairment: mild improvement in threshold performances-results from a randomized controlled study. J Neurotrauma. 2018;
23.
Zurück zum Zitat Bahuleyan B, Singh S. Olfactory memory impairment in neurodegenerative diseases. J Clin Diagnostic Res. 2012;6:1437–41. Bahuleyan B, Singh S. Olfactory memory impairment in neurodegenerative diseases. J Clin Diagnostic Res. 2012;6:1437–41.
24.
Zurück zum Zitat Doty RL. Olfaction in Parkinson’s diseases and related disorders. Neurobiol Dis. 2012;46:527–52.PubMedCrossRef Doty RL. Olfaction in Parkinson’s diseases and related disorders. Neurobiol Dis. 2012;46:527–52.PubMedCrossRef
25.
Zurück zum Zitat Hummel T, Whitcroft KL, Andrews P, et al. Position paper on olfactory dysfunction. Rhinol Suppl. 2017;54:1–30.PubMed Hummel T, Whitcroft KL, Andrews P, et al. Position paper on olfactory dysfunction. Rhinol Suppl. 2017;54:1–30.PubMed
26.
Zurück zum Zitat Morley JF, Duda JE. Olfaction as a biomarker in Parkinson’s disease. Biomark Med. 2010;4:661–70.PubMedCrossRef Morley JF, Duda JE. Olfaction as a biomarker in Parkinson’s disease. Biomark Med. 2010;4:661–70.PubMedCrossRef
27.
Zurück zum Zitat Bowman GL. Biomarkers for early detection of Parkinson’s disease: a scent of consistency with olfactory dysfunction. Neurology. 2017;89:1432–4.PubMedCrossRef Bowman GL. Biomarkers for early detection of Parkinson’s disease: a scent of consistency with olfactory dysfunction. Neurology. 2017;89:1432–4.PubMedCrossRef
28.
29.
Zurück zum Zitat Krismer F, Pinter B, Mueller C, et al. Sniffing the diagnosis: olfactory testing in neurodegenerative parkinsonism. Parkinsonism Relat Disord. 2017;35:36–41.PubMedCrossRef Krismer F, Pinter B, Mueller C, et al. Sniffing the diagnosis: olfactory testing in neurodegenerative parkinsonism. Parkinsonism Relat Disord. 2017;35:36–41.PubMedCrossRef
30.
Zurück zum Zitat Morley JF, Cohen A, Silveira-Moriyama L, et al. Optimizing olfactory testing for the diagnosis of Parkinson’s disease: item analysis of the university of Pennsylvania smell identification test. NPJ Parkinsonism Dis. 2018;4:2.CrossRef Morley JF, Cohen A, Silveira-Moriyama L, et al. Optimizing olfactory testing for the diagnosis of Parkinson’s disease: item analysis of the university of Pennsylvania smell identification test. NPJ Parkinsonism Dis. 2018;4:2.CrossRef
31.
Zurück zum Zitat Tabert MH, Liu X, Doty RL, Serby M, Zamora D, Pelton GH, et al. A 10-item smell identification scale related to risk for Alzheimer’s disease. Ann Neurol. 2005;58:155–60.PubMedCrossRef Tabert MH, Liu X, Doty RL, Serby M, Zamora D, Pelton GH, et al. A 10-item smell identification scale related to risk for Alzheimer’s disease. Ann Neurol. 2005;58:155–60.PubMedCrossRef
32.
Zurück zum Zitat Growdon ME, Schultz AP, Dagley AS, Amarigilio RE, Hedden T, Rentz DM, et al. Odor identification and Alzheimer disease biomarkers in clinically normal elderly. Neurology. 2015;84:2153–60.PubMedPubMedCentralCrossRef Growdon ME, Schultz AP, Dagley AS, Amarigilio RE, Hedden T, Rentz DM, et al. Odor identification and Alzheimer disease biomarkers in clinically normal elderly. Neurology. 2015;84:2153–60.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Lafaille-Magnan ME, Poirier J, Etienne P, Tremblay-Mercier J, Frenette J, Rosa-Neto P, et al. PREVENT-AD Research Group. Odor identification as a biomarker of preclinical AD in older adults at risk. Neurology. 2017;89:327–35.PubMedPubMedCentralCrossRef Lafaille-Magnan ME, Poirier J, Etienne P, Tremblay-Mercier J, Frenette J, Rosa-Neto P, et al. PREVENT-AD Research Group. Odor identification as a biomarker of preclinical AD in older adults at risk. Neurology. 2017;89:327–35.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Woodward MR, Amrutkar CV, Ahah HC, Benedict RH, Rajakrishnan S, Doody RS, et al. Validation of olfactory deficit as a biomarker of Alzheimer disease. Neurol Clin Pract. 2017;7:5–14.PubMedPubMedCentralCrossRef Woodward MR, Amrutkar CV, Ahah HC, Benedict RH, Rajakrishnan S, Doody RS, et al. Validation of olfactory deficit as a biomarker of Alzheimer disease. Neurol Clin Pract. 2017;7:5–14.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat McShane RH, Nagy Z, Esiri MM, King E, Joachim C, Sulivan N, et al. Anosmia in dementia is associated with Lewy bodies rather than Alzheimer’s pathology. J Neurol Neurosurg Psychiatry. 2001;70:739–43.PubMedPubMedCentralCrossRef McShane RH, Nagy Z, Esiri MM, King E, Joachim C, Sulivan N, et al. Anosmia in dementia is associated with Lewy bodies rather than Alzheimer’s pathology. J Neurol Neurosurg Psychiatry. 2001;70:739–43.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat McKeith I, Taylor JP, Thomas A, Donaghy P, Kane J. Revisiting DLB diagnosis: a consideration of prodromal DLB and of the diagnostic overlap with Alzheimer disease. J Geriatr Psychiatry Neurol. 2016;29:249–53.PubMedCrossRef McKeith I, Taylor JP, Thomas A, Donaghy P, Kane J. Revisiting DLB diagnosis: a consideration of prodromal DLB and of the diagnostic overlap with Alzheimer disease. J Geriatr Psychiatry Neurol. 2016;29:249–53.PubMedCrossRef
37.
Zurück zum Zitat Mahlknecht P, Iranzo A, Högi B, et al. Olfactory dysfunction predicts early transition to a Lewy body disease in idiopathic RBD. Neurology. 2015;84:654–8.PubMedCrossRef Mahlknecht P, Iranzo A, Högi B, et al. Olfactory dysfunction predicts early transition to a Lewy body disease in idiopathic RBD. Neurology. 2015;84:654–8.PubMedCrossRef
39.
Zurück zum Zitat Misiak M, Hipolito MM, Ressom HW, Obisesan TO, Manaye KF, Nwlia EA. Apo E4 alleles and impaired olfaction as predictors of Alzheimer’s disease. Clin Exp Psychol. 2017;3:169.PubMedPubMedCentralCrossRef Misiak M, Hipolito MM, Ressom HW, Obisesan TO, Manaye KF, Nwlia EA. Apo E4 alleles and impaired olfaction as predictors of Alzheimer’s disease. Clin Exp Psychol. 2017;3:169.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Kay RB, Meyer EA, Illig KR, Brunjes PC. Spatial distribution of neural activity in the anterior olfactory nucleus evoked by odor and electrical stimulation. J Comp Neurol. 2011;519:277–89.PubMedPubMedCentralCrossRef Kay RB, Meyer EA, Illig KR, Brunjes PC. Spatial distribution of neural activity in the anterior olfactory nucleus evoked by odor and electrical stimulation. J Comp Neurol. 2011;519:277–89.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Doty RL, Philip S, Reddy K, Kerr KL. Influences of antihypertensive and antihyperlipidemic drugs on the senses of taste and smell: a review. J Hypertens. 2003;21:1805–13.PubMedCrossRef Doty RL, Philip S, Reddy K, Kerr KL. Influences of antihypertensive and antihyperlipidemic drugs on the senses of taste and smell: a review. J Hypertens. 2003;21:1805–13.PubMedCrossRef
43.
Zurück zum Zitat Buck L, Axel R. A novel multigene family may encode odorant receptors: a molecular basis for odor recognition. Cell. 1991;65:175–87.PubMedCrossRef Buck L, Axel R. A novel multigene family may encode odorant receptors: a molecular basis for odor recognition. Cell. 1991;65:175–87.PubMedCrossRef
44.
Zurück zum Zitat Masurkar AV, Devanand DP. Olfactory dysfunction in the elderly: basic circuitry and alterations with normal aging and Alzheimer’s disease. Curr Geriatr Rep. 2014;3:91–100.PubMedPubMedCentralCrossRef Masurkar AV, Devanand DP. Olfactory dysfunction in the elderly: basic circuitry and alterations with normal aging and Alzheimer’s disease. Curr Geriatr Rep. 2014;3:91–100.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Verhagen JV, Wesson DW, Netoff TI, White JA, Wachowiak M. Sniffing controls and adaptive filter of sensory input to the olfactory bulb. Nat Neurosci. 2007;10:631–9.PubMedCrossRef Verhagen JV, Wesson DW, Netoff TI, White JA, Wachowiak M. Sniffing controls and adaptive filter of sensory input to the olfactory bulb. Nat Neurosci. 2007;10:631–9.PubMedCrossRef
46.
Zurück zum Zitat Carey RM, Wachowiak M. Effect of sniffing on the temporal structure of mitral/tufted cell output from the olfactory bulb. J Neurosci. 2011;31:10615–26.PubMedPubMedCentralCrossRef Carey RM, Wachowiak M. Effect of sniffing on the temporal structure of mitral/tufted cell output from the olfactory bulb. J Neurosci. 2011;31:10615–26.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Chess A, Simon I, Cedar H, Axel R. Allelic inactivation regulates olfactory receptor gene expression. Cell. 1994;78:823–34.PubMedCrossRef Chess A, Simon I, Cedar H, Axel R. Allelic inactivation regulates olfactory receptor gene expression. Cell. 1994;78:823–34.PubMedCrossRef
48.
Zurück zum Zitat Shykind BM, Rohani SC, O’Donnell S, Nemes A, Mendelsohn M, Sun Y, et al. Gene switching and the stability of odorant receptor gene choice. Cell. 2004;117:801–15.PubMedCrossRef Shykind BM, Rohani SC, O’Donnell S, Nemes A, Mendelsohn M, Sun Y, et al. Gene switching and the stability of odorant receptor gene choice. Cell. 2004;117:801–15.PubMedCrossRef
49.
Zurück zum Zitat Mombaerts P, Wang F, Dulac C, Chao SK, Nemes A, Mendelsohn M, et al. Visualizing an olfactory sensory map. Cell. 1996;87:675–86.PubMedCrossRef Mombaerts P, Wang F, Dulac C, Chao SK, Nemes A, Mendelsohn M, et al. Visualizing an olfactory sensory map. Cell. 1996;87:675–86.PubMedCrossRef
50.
Zurück zum Zitat Liu A, Savya S, Urban NN. Early odorant exposure increases the number of mitral cells associated with a single glomerulus. J Neurosci. 2016;36:11646–53.PubMedPubMedCentralCrossRef Liu A, Savya S, Urban NN. Early odorant exposure increases the number of mitral cells associated with a single glomerulus. J Neurosci. 2016;36:11646–53.PubMedPubMedCentralCrossRef
51.
52.
Zurück zum Zitat Dunkel A, Steinhaus M, Kotthoff M, Nowak B, Krautwurst D, Schieberle P, et al. Nature’s chemical signatures in human olfaction: a foodborne perspective for future biotechnology. Angew Chem Int Ed Engl. 2014;53:7124–43.PubMedCrossRef Dunkel A, Steinhaus M, Kotthoff M, Nowak B, Krautwurst D, Schieberle P, et al. Nature’s chemical signatures in human olfaction: a foodborne perspective for future biotechnology. Angew Chem Int Ed Engl. 2014;53:7124–43.PubMedCrossRef
54.
Zurück zum Zitat Nagayama S, Takakhashi YK, Yoshihara Y, Mori K. Mitral and tufted cells differ in the decoding manner of odor maps in the rat olfactory bulb. J Neurophysiol. 2004;91:2532–40.PubMedCrossRef Nagayama S, Takakhashi YK, Yoshihara Y, Mori K. Mitral and tufted cells differ in the decoding manner of odor maps in the rat olfactory bulb. J Neurophysiol. 2004;91:2532–40.PubMedCrossRef
55.
Zurück zum Zitat Linster C, Fontanini A. Functional neuromodulation of chemosensation in vertebrates. Curr Opin Neurobiol. 2014;29:82–7.PubMedCrossRef Linster C, Fontanini A. Functional neuromodulation of chemosensation in vertebrates. Curr Opin Neurobiol. 2014;29:82–7.PubMedCrossRef
56.
Zurück zum Zitat Bendahmane M, Cameron M, Ennis M, Fletcher ML. Increased olfactory bulb acetycholine bi-directionally modulates glomerular odor sensitivity. Scientifc Reports. 2016;6:25808.CrossRef Bendahmane M, Cameron M, Ennis M, Fletcher ML. Increased olfactory bulb acetycholine bi-directionally modulates glomerular odor sensitivity. Scientifc Reports. 2016;6:25808.CrossRef
57.
Zurück zum Zitat Huang Z, Thiebaud N, Fadool DA. Differential serotonergic modulation across the main and accessory olfactory bulbs. J Physiol. 2017;595:3515–33.PubMedPubMedCentralCrossRef Huang Z, Thiebaud N, Fadool DA. Differential serotonergic modulation across the main and accessory olfactory bulbs. J Physiol. 2017;595:3515–33.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Höglinger GU, Rizk P, Muriel MP, Duyckaerts C, Oertel WH, Caille I, et al. Dopamine depletion impairs precursor cell proliferation in Parkinson’s disease. Nature Neurosci. 2004;7:726–35.PubMedCrossRef Höglinger GU, Rizk P, Muriel MP, Duyckaerts C, Oertel WH, Caille I, et al. Dopamine depletion impairs precursor cell proliferation in Parkinson’s disease. Nature Neurosci. 2004;7:726–35.PubMedCrossRef
59.
Zurück zum Zitat Grier BD, Belluscio L, Cheetham CE. Olfactory sensory activity modulates microglial-neuronal interactions during dopaminergic cell loss in the olfactory bulb. Front Cell Neurosci. 2016;10:178.PubMedPubMedCentralCrossRef Grier BD, Belluscio L, Cheetham CE. Olfactory sensory activity modulates microglial-neuronal interactions during dopaminergic cell loss in the olfactory bulb. Front Cell Neurosci. 2016;10:178.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Höglinger GU, Alvarez-Fischer D, Arias-Carrión I, Djufri M, Windolph A, Keber U, et al. A new dopaminergic nigro-olfactory projection. Acta Neuropathol. 2015;130:333–48.PubMedCrossRef Höglinger GU, Alvarez-Fischer D, Arias-Carrión I, Djufri M, Windolph A, Keber U, et al. A new dopaminergic nigro-olfactory projection. Acta Neuropathol. 2015;130:333–48.PubMedCrossRef
61.
Zurück zum Zitat Alvarez-Buylla A, Kohwi M, Nguyen TM, Merkle FT. The heterogeneity of adult neural stem cells and the emerging complexity of their niche. Cold Spring Harb Symp Quant Biol. 2008;73:357–65.PubMedCrossRef Alvarez-Buylla A, Kohwi M, Nguyen TM, Merkle FT. The heterogeneity of adult neural stem cells and the emerging complexity of their niche. Cold Spring Harb Symp Quant Biol. 2008;73:357–65.PubMedCrossRef
64.
Zurück zum Zitat Ubeda-Bañón I, Saiz-Sanchez D, de la Rosa-Prieto C, Martinez-Marcos A. α-Synuclein in the olfactory system in Parkinsons’ disease: role of neural connections on spreading pathology. Brain Struct Funct. 2014;19:1513–26. Ubeda-Bañón I, Saiz-Sanchez D, de la Rosa-Prieto C, Martinez-Marcos A. α-Synuclein in the olfactory system in Parkinsons’ disease: role of neural connections on spreading pathology. Brain Struct Funct. 2014;19:1513–26.
66.
Zurück zum Zitat Doty RL, Deems DA, Stellar S. Olfactory dysfunction in parkinsonism: a general deficit unrelated to neurologic signs, disease stage, or disease duration. Neurology. 1988;38:1237–44.PubMedCrossRef Doty RL, Deems DA, Stellar S. Olfactory dysfunction in parkinsonism: a general deficit unrelated to neurologic signs, disease stage, or disease duration. Neurology. 1988;38:1237–44.PubMedCrossRef
67.
Zurück zum Zitat Silveira-Moriyama L, Sirisena D, Gamage P, Gamage R, de Silva R, Lees AJ. Adapting the sniffin’ sticks to diagnose Parkinson’s disease in Sri Lanka. Mov Disord. 2009;24:1229–33.PubMedCrossRef Silveira-Moriyama L, Sirisena D, Gamage P, Gamage R, de Silva R, Lees AJ. Adapting the sniffin’ sticks to diagnose Parkinson’s disease in Sri Lanka. Mov Disord. 2009;24:1229–33.PubMedCrossRef
68.
Zurück zum Zitat Kobal G, Hummel T, Sekinger B, Baez S, Roscher S, Wolf S. “Sniffin’sticks”: screening of olfactory performance. Rhinology. 1996;34:222–6.PubMed Kobal G, Hummel T, Sekinger B, Baez S, Roscher S, Wolf S. “Sniffin’sticks”: screening of olfactory performance. Rhinology. 1996;34:222–6.PubMed
69.
Zurück zum Zitat Hummel T, Sekinger B, Wolf SR, Pauli E, Kobal G. “Sniffin’ sticks”: olfactory performance assessed by the combined testing of odor identification, odor discrimination and olfactory threshold. Chem Senses. 1997;22:39–52.PubMedCrossRef Hummel T, Sekinger B, Wolf SR, Pauli E, Kobal G. “Sniffin’ sticks”: olfactory performance assessed by the combined testing of odor identification, odor discrimination and olfactory threshold. Chem Senses. 1997;22:39–52.PubMedCrossRef
70.
Zurück zum Zitat Haehner A, Tosch C, Wolz M, Klingelhoefer L, fauser M, Storch A, et al. Olfactory training in patients with Parkinson’s disease. PLoS One. 2013;8:e61680.PubMedPubMedCentralCrossRef Haehner A, Tosch C, Wolz M, Klingelhoefer L, fauser M, Storch A, et al. Olfactory training in patients with Parkinson’s disease. PLoS One. 2013;8:e61680.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Doty RL, Marcus A, Lee WW. Development of the 12-item cross-cultural smell identification test (CC-SIT). Laryngoscope. 1996;106:353–6.PubMedCrossRef Doty RL, Marcus A, Lee WW. Development of the 12-item cross-cultural smell identification test (CC-SIT). Laryngoscope. 1996;106:353–6.PubMedCrossRef
72.
Zurück zum Zitat Rodriguez-Violante M, Glonzalez-Latapi P, Camacho-Ordoñez A, Martínez-Ramírez D, Morales-Briceño H, Cervantes-Arriaga A. Low specificity and sensitivity of smell identification testing for the diagnosis of Parkinson’s disease. Arq Neuropsiquiatry. 2014;72:33–7.CrossRef Rodriguez-Violante M, Glonzalez-Latapi P, Camacho-Ordoñez A, Martínez-Ramírez D, Morales-Briceño H, Cervantes-Arriaga A. Low specificity and sensitivity of smell identification testing for the diagnosis of Parkinson’s disease. Arq Neuropsiquiatry. 2014;72:33–7.CrossRef
73.
Zurück zum Zitat Cardesin A, Alobid I, Benítez P, Sierra E, de Haro J, Bernal-Sprekelsen M, et al. Barcelona Smell Test-24 (BAST-24): validation and smell characteristics in the healthy Spanish population. Rhinology. 2006;44:83–9.PubMed Cardesin A, Alobid I, Benítez P, Sierra E, de Haro J, Bernal-Sprekelsen M, et al. Barcelona Smell Test-24 (BAST-24): validation and smell characteristics in the healthy Spanish population. Rhinology. 2006;44:83–9.PubMed
74.
Zurück zum Zitat Iijima M, Kobayakawa T, Saito S, Osawa M, Tsutsumo Y, Hashimoto S, et al. Smell identification in Japanese Parkinson’s disease patients: using the odor stick identification test for Japanese subjects. Intern Med. 2008;47:1887–92.PubMedCrossRef Iijima M, Kobayakawa T, Saito S, Osawa M, Tsutsumo Y, Hashimoto S, et al. Smell identification in Japanese Parkinson’s disease patients: using the odor stick identification test for Japanese subjects. Intern Med. 2008;47:1887–92.PubMedCrossRef
75.
Zurück zum Zitat Maremmani C, Rossi G, Tambasco N, et al. The validity and reliability of the Italian olfactory identification test (IOIT) in healthy subjects and in Parkinson’s disease patients. Parkinsonism Relat Disord. 2012;18:788–93.PubMedCrossRef Maremmani C, Rossi G, Tambasco N, et al. The validity and reliability of the Italian olfactory identification test (IOIT) in healthy subjects and in Parkinson’s disease patients. Parkinsonism Relat Disord. 2012;18:788–93.PubMedCrossRef
76.
Zurück zum Zitat Mariño-Sánchez F, Valls-Mateus M, Haag O, Alobid I, Busquet J, Mullol J. Smell loss is associated with severe and uncontrolled disease in children and adolescents with persisting allergic rhinitis. J Allergy Clin Immunol Pract. 2018. Mariño-Sánchez F, Valls-Mateus M, Haag O, Alobid I, Busquet J, Mullol J. Smell loss is associated with severe and uncontrolled disease in children and adolescents with persisting allergic rhinitis. J Allergy Clin Immunol Pract. 2018.
77.
Zurück zum Zitat Vilas D, Quintana M, Pont-Sunyer C, et al. Olfactory characterization of idiopathic Parkinson’s disease and LRRK2 associated parkinsonism: a practical approach (submitted). Vilas D, Quintana M, Pont-Sunyer C, et al. Olfactory characterization of idiopathic Parkinson’s disease and LRRK2 associated parkinsonism: a practical approach (submitted).
78.
Zurück zum Zitat Liu G, Zong G, Doty RL, Sun Q. Prevalence and risk factors of taste and smell impairment in a nationwide representative sample of the US population: a cross-sectional study. BMJ Open. 2016;6:e013246.PubMedPubMedCentralCrossRef Liu G, Zong G, Doty RL, Sun Q. Prevalence and risk factors of taste and smell impairment in a nationwide representative sample of the US population: a cross-sectional study. BMJ Open. 2016;6:e013246.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Gros A, Manera V, de March CA, et al. Olfactory disturbances in ageing with and without dementia: towards new diagnostic tools. J Laryngol Otol. 2017;131:572–9.PubMedCrossRef Gros A, Manera V, de March CA, et al. Olfactory disturbances in ageing with and without dementia: towards new diagnostic tools. J Laryngol Otol. 2017;131:572–9.PubMedCrossRef
80.
Zurück zum Zitat Murphy C, Gilmore MM, Seery CS, Salmon DP, Lasker BR. Olfactory thresholds are associated with degree of dementia in Alzheimer’s disease. Neurobiol Aging. 1990;11:465–9.PubMedCrossRef Murphy C, Gilmore MM, Seery CS, Salmon DP, Lasker BR. Olfactory thresholds are associated with degree of dementia in Alzheimer’s disease. Neurobiol Aging. 1990;11:465–9.PubMedCrossRef
81.
82.
Zurück zum Zitat Doty RL, Sharman P, Applebaum SL, Giberson R, Siksorski L, Rosenberg L. Smell identification ability: changes with age. Science. 1984;226:1441–3.PubMedCrossRef Doty RL, Sharman P, Applebaum SL, Giberson R, Siksorski L, Rosenberg L. Smell identification ability: changes with age. Science. 1984;226:1441–3.PubMedCrossRef
83.
Zurück zum Zitat Choudhury ES, Moberg P, Doty RL. Influences of age and sex on a microencapsulated odor memory test. Chem Senses. 2003;28:799–805.PubMedCrossRef Choudhury ES, Moberg P, Doty RL. Influences of age and sex on a microencapsulated odor memory test. Chem Senses. 2003;28:799–805.PubMedCrossRef
84.
Zurück zum Zitat Oliveira-Pinto AV, Santos RM, Coutinho RA, et al. Sexual dimorphism in the human olfactory bulb: females have more neurons and glial cells than males. PLoS Ones. 2014;9:e111733.CrossRef Oliveira-Pinto AV, Santos RM, Coutinho RA, et al. Sexual dimorphism in the human olfactory bulb: females have more neurons and glial cells than males. PLoS Ones. 2014;9:e111733.CrossRef
85.
Zurück zum Zitat Xydakis MS, Belluscio L. Detection of neurodegenerative disease using olfaction. Lancet Neurol. 2017;16:415–6.PubMedCrossRef Xydakis MS, Belluscio L. Detection of neurodegenerative disease using olfaction. Lancet Neurol. 2017;16:415–6.PubMedCrossRef
86.
Zurück zum Zitat Hummel T, Heilmann S, Murphy C. Age-related changes in chemosensory functions. In: Rouby C, et al., editors. Olfaction, taste and cognition. New York: Cambridge University Press; 2002. p. 451–6. Hummel T, Heilmann S, Murphy C. Age-related changes in chemosensory functions. In: Rouby C, et al., editors. Olfaction, taste and cognition. New York: Cambridge University Press; 2002. p. 451–6.
87.
Zurück zum Zitat Murphy C, Nordin S, Acosta L. Odor learning, recall, and recognition memory in young and elderly adults. Neuropsychology. 1997;11:126–37.PubMedCrossRef Murphy C, Nordin S, Acosta L. Odor learning, recall, and recognition memory in young and elderly adults. Neuropsychology. 1997;11:126–37.PubMedCrossRef
88.
Zurück zum Zitat Suzuki Y, Critchley HD, Suckling J, et al. Functional magnetic resonance imaging of odor identification: the effect of aging. J Gerontol A Biol Sci Med Sci. 2001;56:M756–60.PubMedCrossRef Suzuki Y, Critchley HD, Suckling J, et al. Functional magnetic resonance imaging of odor identification: the effect of aging. J Gerontol A Biol Sci Med Sci. 2001;56:M756–60.PubMedCrossRef
89.
Zurück zum Zitat Cerf-Ducastel B, Murphy C. fMRI brain activation in response to odors is reduced in primary olfactory areas of elderly subjects. Brain Res. 2003;986:39–53.PubMedCrossRef Cerf-Ducastel B, Murphy C. fMRI brain activation in response to odors is reduced in primary olfactory areas of elderly subjects. Brain Res. 2003;986:39–53.PubMedCrossRef
90.
Zurück zum Zitat Ferdon S, Murphy C. The cerebellum and olfaction in the aging brain: a functional magnetic resonance imaging study. Neuroimage. 2003;20:12–21.PubMedCrossRef Ferdon S, Murphy C. The cerebellum and olfaction in the aging brain: a functional magnetic resonance imaging study. Neuroimage. 2003;20:12–21.PubMedCrossRef
91.
Zurück zum Zitat Rawson NE. Olfactory loss in aging. Sci Aging Knowl Environ. 2006;5:pe6. Rawson NE. Olfactory loss in aging. Sci Aging Knowl Environ. 2006;5:pe6.
93.
Zurück zum Zitat Mobley AS, Rodriguez-Gil DJ, Imamura F, Greer CA. Aging in the olfactory system. Trends Neurosci. 2014;37:77–84.PubMedCrossRef Mobley AS, Rodriguez-Gil DJ, Imamura F, Greer CA. Aging in the olfactory system. Trends Neurosci. 2014;37:77–84.PubMedCrossRef
94.
Zurück zum Zitat Paik SI, Lehman MN, Seiden AM, Duncan HJ, Smith DV. Human olfactory biopsy. The influence of age and receptor distribution. Arch Otolaryngol Head Neck Surg. 1992;118:731–8.PubMedCrossRef Paik SI, Lehman MN, Seiden AM, Duncan HJ, Smith DV. Human olfactory biopsy. The influence of age and receptor distribution. Arch Otolaryngol Head Neck Surg. 1992;118:731–8.PubMedCrossRef
95.
Zurück zum Zitat Rosli Y, Brecjenridge LJ, Smith RA. An ultrastrucutral study of age-related changes in mouse olfactory epithelium. J Electron Microsc. 1999;48:77–84.CrossRef Rosli Y, Brecjenridge LJ, Smith RA. An ultrastrucutral study of age-related changes in mouse olfactory epithelium. J Electron Microsc. 1999;48:77–84.CrossRef
96.
Zurück zum Zitat Ueha R, Schichino S, Ueha S, Kondo K, Kikuta S, Nishijima H, et al. Reduction of proliferating olfactory cells and low expression of extracellular matrix genes are hallmarks of the aged olfactory mucosa. Front Aging Neurosci. 2018;10:86.PubMedPubMedCentralCrossRef Ueha R, Schichino S, Ueha S, Kondo K, Kikuta S, Nishijima H, et al. Reduction of proliferating olfactory cells and low expression of extracellular matrix genes are hallmarks of the aged olfactory mucosa. Front Aging Neurosci. 2018;10:86.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Buschhüter D, Smitka M, Puschmann S, Gerber JC, Abolmaali ND, Hummel T. Correlation between olfactory bulb volume and olfactory function. Neuroimage. 2008;42:498–502.PubMedCrossRef Buschhüter D, Smitka M, Puschmann S, Gerber JC, Abolmaali ND, Hummel T. Correlation between olfactory bulb volume and olfactory function. Neuroimage. 2008;42:498–502.PubMedCrossRef
98.
Zurück zum Zitat Rombaux P, Duprez T, Hummel T. Olfactory bulb volume in the clinical assessment of olfactory dysfunction. Rhinology. 2009;47:3–9.PubMed Rombaux P, Duprez T, Hummel T. Olfactory bulb volume in the clinical assessment of olfactory dysfunction. Rhinology. 2009;47:3–9.PubMed
99.
Zurück zum Zitat Rombaux P, Huart C, Deggouj N, Duprez T, Hummel T. Prognostic value of olfactory bulb volume measurement for recovery in postinfectious and posttraumatic olfactory loss. Otolaryngol Head Neck Surg. 2012;147:1136–41.PubMedCrossRef Rombaux P, Huart C, Deggouj N, Duprez T, Hummel T. Prognostic value of olfactory bulb volume measurement for recovery in postinfectious and posttraumatic olfactory loss. Otolaryngol Head Neck Surg. 2012;147:1136–41.PubMedCrossRef
100.
Zurück zum Zitat Paschen L, Schmidt N, Wolff S, Cnyrim C, van Eimeren T, Zeuner KE, et al. The olfactory bulb volume in patients with idiopathic Parkinson’s disease. Eur J Neurol. 2015;22:1068–73.PubMedCrossRef Paschen L, Schmidt N, Wolff S, Cnyrim C, van Eimeren T, Zeuner KE, et al. The olfactory bulb volume in patients with idiopathic Parkinson’s disease. Eur J Neurol. 2015;22:1068–73.PubMedCrossRef
101.
Zurück zum Zitat Mazal PP, Haehner A, Hummel T. Relation of the volume of the olfactory bulb to psychophysical measures of olfactory function. Eur Arch Otorhinolaryngol. 2016;273:1–7.PubMedCrossRef Mazal PP, Haehner A, Hummel T. Relation of the volume of the olfactory bulb to psychophysical measures of olfactory function. Eur Arch Otorhinolaryngol. 2016;273:1–7.PubMedCrossRef
102.
Zurück zum Zitat •• Marin C, Laxe S, Langdon C, Berenguer J, Lehrer E, Mariño-Sánchez F, et al. Olfactory function in an excitotoxic model for secondary neuronal degeneration: role of dopaminergic interneurons. Neuroscience. 2017;364:28–44. Preclinical evidence of the lack of correlation between olfactory dysfunction and recovery and the olfactory bulbs volume. PubMedCrossRef •• Marin C, Laxe S, Langdon C, Berenguer J, Lehrer E, Mariño-Sánchez F, et al. Olfactory function in an excitotoxic model for secondary neuronal degeneration: role of dopaminergic interneurons. Neuroscience. 2017;364:28–44. Preclinical evidence of the lack of correlation between olfactory dysfunction and recovery and the olfactory bulbs volume. PubMedCrossRef
103.
Zurück zum Zitat Segura B, Baggio HC, Sola E, Palacios EM, Vendrell P, Bargalló N, et al. Neuroanatomical correlates of olfactory loss in normal aged subjects. Behav Brain Res. 2013;246:148–53.PubMedCrossRef Segura B, Baggio HC, Sola E, Palacios EM, Vendrell P, Bargalló N, et al. Neuroanatomical correlates of olfactory loss in normal aged subjects. Behav Brain Res. 2013;246:148–53.PubMedCrossRef
104.
Zurück zum Zitat Adjei S, Houck AL, Ma K, Wesson DW. Age-dependent alterations in the number, volume, and localization of islands of Calleja within the olfactory tubercle. Neurobiol Aging. 2013;34:2676–82.PubMedCrossRef Adjei S, Houck AL, Ma K, Wesson DW. Age-dependent alterations in the number, volume, and localization of islands of Calleja within the olfactory tubercle. Neurobiol Aging. 2013;34:2676–82.PubMedCrossRef
105.
Zurück zum Zitat Wilson RS, Schneider JA, Arnold SE, Tang Y, Boyle PA, Benner DA. Olfactory identification and incidence of mild cognitive impairment in older age. Arch Gen Psychiatry. 2007;64:802–8.PubMedCrossRef Wilson RS, Schneider JA, Arnold SE, Tang Y, Boyle PA, Benner DA. Olfactory identification and incidence of mild cognitive impairment in older age. Arch Gen Psychiatry. 2007;64:802–8.PubMedCrossRef
106.
Zurück zum Zitat Gopinath B, Sue CM, Kifley A, Mitchell P. The association between olfactory impairment and total mortality in older adults. J Gerontol A Biol Sci Med Sci. 2012;67:204–9.PubMedCrossRef Gopinath B, Sue CM, Kifley A, Mitchell P. The association between olfactory impairment and total mortality in older adults. J Gerontol A Biol Sci Med Sci. 2012;67:204–9.PubMedCrossRef
107.
Zurück zum Zitat Devanand DP, Lee S, Manly J, Andrews H, Schupf N, Masurkar A, et al. Olfactory identification deficits and increased mortality in the community. Ann Neurol. 2015;78:401–11.PubMedPubMedCentralCrossRef Devanand DP, Lee S, Manly J, Andrews H, Schupf N, Masurkar A, et al. Olfactory identification deficits and increased mortality in the community. Ann Neurol. 2015;78:401–11.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Almkvist O, Berglund B, Nordin S. Odor detectability in successfully aged elderly and young adults. Reports from the Dept of Psychology, Stockholm University. 1992;744:1–12. Almkvist O, Berglund B, Nordin S. Odor detectability in successfully aged elderly and young adults. Reports from the Dept of Psychology, Stockholm University. 1992;744:1–12.
109.
Zurück zum Zitat Mackay-Sim A, Johnston AN, Owen C, Burne TH. Olfactory ability in the healthy population: reassessing presbyosmia. Chem Senses. 2006;31:763–71.PubMedCrossRef Mackay-Sim A, Johnston AN, Owen C, Burne TH. Olfactory ability in the healthy population: reassessing presbyosmia. Chem Senses. 2006;31:763–71.PubMedCrossRef
110.
Zurück zum Zitat Schubert CR, Carmichael LL, Murphy C, Klein BE, Kelin R, Cruickshanks KJ. Olfaction and the 5-year incidence of cognitive impairment in an epidemiological study of older adults. J Am Geriatr Soc. 2008;56:1517–21.PubMedPubMedCentralCrossRef Schubert CR, Carmichael LL, Murphy C, Klein BE, Kelin R, Cruickshanks KJ. Olfaction and the 5-year incidence of cognitive impairment in an epidemiological study of older adults. J Am Geriatr Soc. 2008;56:1517–21.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Kreisi WC, Jin P, Lee S, et al. Odor identification ability predicts PET amyloid status and memory decline in older adults. J Alzheimers Dis. 2018;62:1759–66.CrossRef Kreisi WC, Jin P, Lee S, et al. Odor identification ability predicts PET amyloid status and memory decline in older adults. J Alzheimers Dis. 2018;62:1759–66.CrossRef
112.
Zurück zum Zitat Kovacs T. Mechanisms of olfactory dysfunction in aging and neurodegenerative disorders. Ageing Res Rev. 2004;3:215–32.PubMedCrossRef Kovacs T. Mechanisms of olfactory dysfunction in aging and neurodegenerative disorders. Ageing Res Rev. 2004;3:215–32.PubMedCrossRef
113.
Zurück zum Zitat Gerkin RC, Adler CH, Hentz JG, et al. Improved diagnosis of Parkinson’s disease from a detailed olfactory phenotype. Ann Clin Transl Neurol. 2017;4:714–21.PubMedPubMedCentralCrossRef Gerkin RC, Adler CH, Hentz JG, et al. Improved diagnosis of Parkinson’s disease from a detailed olfactory phenotype. Ann Clin Transl Neurol. 2017;4:714–21.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Godoy MD, Voegels RL, de Pinna F, Imamura R, Farfel JM. Olfaction in neurologic and neurodegenerative diseases: a literature review. Int Arch Otorhinolaryngol. 2015;19:176–9.PubMed Godoy MD, Voegels RL, de Pinna F, Imamura R, Farfel JM. Olfaction in neurologic and neurodegenerative diseases: a literature review. Int Arch Otorhinolaryngol. 2015;19:176–9.PubMed
115.
Zurück zum Zitat Hawkes C. Olfaction in neurodegenerative disorder. Adv Otorhinolaryngol. 2006;63:133–51.PubMed Hawkes C. Olfaction in neurodegenerative disorder. Adv Otorhinolaryngol. 2006;63:133–51.PubMed
116.
Zurück zum Zitat Attems J, Walker K, Jellinger KA. Olfactory bulb involvement in neurodegenerative diseases. Acta Neuropathol. 2014;127:459–75.PubMedCrossRef Attems J, Walker K, Jellinger KA. Olfactory bulb involvement in neurodegenerative diseases. Acta Neuropathol. 2014;127:459–75.PubMedCrossRef
117.
Zurück zum Zitat Driver-Dunckley E, Adler CH, Hentz JG, Dugger BN, Shill HA, Caviness JN, et al. Olfactory dysfunction in incidental Lewy body disease and Parkinson’s disease. Parkinsonism Relat Disord. 2014;20:1260–2.PubMedPubMedCentralCrossRef Driver-Dunckley E, Adler CH, Hentz JG, Dugger BN, Shill HA, Caviness JN, et al. Olfactory dysfunction in incidental Lewy body disease and Parkinson’s disease. Parkinsonism Relat Disord. 2014;20:1260–2.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Velayudhan L. Smell identfication function and Alzheimer’s disease: a selective review. Curr Opin Psychiatry. 2015;28:173–9.PubMed Velayudhan L. Smell identfication function and Alzheimer’s disease: a selective review. Curr Opin Psychiatry. 2015;28:173–9.PubMed
119.
Zurück zum Zitat Huang SF, Chen K, Wu JJ, et al. Odor identification test in idiopathic REM-behavior disorder and Parkinson’s disease in China. PLoS One. 2016;11:e0160199.PubMedPubMedCentralCrossRef Huang SF, Chen K, Wu JJ, et al. Odor identification test in idiopathic REM-behavior disorder and Parkinson’s disease in China. PLoS One. 2016;11:e0160199.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Adler CH. Premotor symptoms and early diagnosis of Parkinson’s disease. Int J Neurosci. 2011;121(Suppl 2):3–8.PubMedCrossRef Adler CH. Premotor symptoms and early diagnosis of Parkinson’s disease. Int J Neurosci. 2011;121(Suppl 2):3–8.PubMedCrossRef
121.
Zurück zum Zitat Hüttenbrink KB, Hummel T, Berg D, Gasser T, Hähner A. Olfactory dysfunction: common in later life and early warning of neurodegenerative disease. Dtsch Arztebl Int. 2013;110:1–7.PubMedPubMedCentral Hüttenbrink KB, Hummel T, Berg D, Gasser T, Hähner A. Olfactory dysfunction: common in later life and early warning of neurodegenerative disease. Dtsch Arztebl Int. 2013;110:1–7.PubMedPubMedCentral
122.
Zurück zum Zitat Braak H, Braak E. Neuropathological staging of Alzheimer-related changes. Acta Neuropathol. 1991;82:239–59.PubMedCrossRef Braak H, Braak E. Neuropathological staging of Alzheimer-related changes. Acta Neuropathol. 1991;82:239–59.PubMedCrossRef
123.
Zurück zum Zitat Braak H, Del Tredici K, Rub U, de Vos RA, Jansen EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.PubMedCrossRef Braak H, Del Tredici K, Rub U, de Vos RA, Jansen EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211.PubMedCrossRef
124.
Zurück zum Zitat Doty RL. The olfactory vector hypothesis of neurodegenerative disease: is it viable? Ann Neurol. 2008;63:7–15.PubMedCrossRef Doty RL. The olfactory vector hypothesis of neurodegenerative disease: is it viable? Ann Neurol. 2008;63:7–15.PubMedCrossRef
125.
Zurück zum Zitat Hobson DE. Asymmetry in parkinsonism, spreading pathogens and the nose. Parkinsonism Relat Disord. 2012;18:1–9.PubMedCrossRef Hobson DE. Asymmetry in parkinsonism, spreading pathogens and the nose. Parkinsonism Relat Disord. 2012;18:1–9.PubMedCrossRef
126.
Zurück zum Zitat Adler CH, Connor DJ, Hentz JG, Sabbagh MN, Caviness JN, Shill HA, et al. Incidental Lewy body disease: clinical comparison to a control cohort. Mov Disord. 2010;25:642–6.PubMedPubMedCentralCrossRef Adler CH, Connor DJ, Hentz JG, Sabbagh MN, Caviness JN, Shill HA, et al. Incidental Lewy body disease: clinical comparison to a control cohort. Mov Disord. 2010;25:642–6.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Beach TG, Adler C, Lue L, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117:613–34.PubMedPubMedCentralCrossRef Beach TG, Adler C, Lue L, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117:613–34.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Jellinger KA. Lewy body-related alpha-synucleopathy in the aged human brain. J Neural Transm. 2004;111:1219–35.PubMedCrossRef Jellinger KA. Lewy body-related alpha-synucleopathy in the aged human brain. J Neural Transm. 2004;111:1219–35.PubMedCrossRef
129.
Zurück zum Zitat Saito Y, Ruberu NN, Sawabe M, Arai T, Kazam H, Hosoi T, et al. Lewy body-related alpha-synucleinopathy in aging. J Neuropathol Exp Neurol. 2004;63:742–9.PubMedCrossRef Saito Y, Ruberu NN, Sawabe M, Arai T, Kazam H, Hosoi T, et al. Lewy body-related alpha-synucleinopathy in aging. J Neuropathol Exp Neurol. 2004;63:742–9.PubMedCrossRef
130.
Zurück zum Zitat Ross GW, Abbott RD, Petrovitch H, et al. Association of olfactory dysfunction with incidental Lewy bodies. Mov Disord. 2006;12:2062–7.CrossRef Ross GW, Abbott RD, Petrovitch H, et al. Association of olfactory dysfunction with incidental Lewy bodies. Mov Disord. 2006;12:2062–7.CrossRef
131.
Zurück zum Zitat Iranzo A, Molinuevo JL, Santamaria J, et al. Rapid-eye-movement sleep behavior disorder as an early marker for a neurodegenerative disorder: a description study. Lancet. 2006;5:572–7.PubMedCrossRef Iranzo A, Molinuevo JL, Santamaria J, et al. Rapid-eye-movement sleep behavior disorder as an early marker for a neurodegenerative disorder: a description study. Lancet. 2006;5:572–7.PubMedCrossRef
132.
Zurück zum Zitat Iranzo A, Tolosa E, Gelpi E, et al. Neurodegenerative disease status and post-mortem pathology in idiopathic rapid-eye movement sleep behavior disorder: and observational cohort study. Lancet Neurol. 2013;12:443–53.PubMedCrossRef Iranzo A, Tolosa E, Gelpi E, et al. Neurodegenerative disease status and post-mortem pathology in idiopathic rapid-eye movement sleep behavior disorder: and observational cohort study. Lancet Neurol. 2013;12:443–53.PubMedCrossRef
133.
Zurück zum Zitat Postuma RB, Gagnon JF, Vendette M, Desjardins C, Montplaisir JY. Olfaction and color vision identify impending neurodegeneration in rapid eye movement sleep behavior disorder. Ann Neurol. 2011;69:811–8.PubMedCrossRef Postuma RB, Gagnon JF, Vendette M, Desjardins C, Montplaisir JY. Olfaction and color vision identify impending neurodegeneration in rapid eye movement sleep behavior disorder. Ann Neurol. 2011;69:811–8.PubMedCrossRef
134.
Zurück zum Zitat Park JW, Kwon DY, Choi JH, Park MH, Yoon HK. Olfactory dysfunctions in drug-naïve Parkinson’s disease with mild cognitive impairment. Parkinsonism Relat Disord. 2018;46:69–73.PubMedCrossRef Park JW, Kwon DY, Choi JH, Park MH, Yoon HK. Olfactory dysfunctions in drug-naïve Parkinson’s disease with mild cognitive impairment. Parkinsonism Relat Disord. 2018;46:69–73.PubMedCrossRef
135.
Zurück zum Zitat Schapira AHV, Chaudhuri KR, Jenner P. Non-motor features of Parkinson’s disease. Nat Rev Neurosci. 2017;18:435–50.PubMedCrossRef Schapira AHV, Chaudhuri KR, Jenner P. Non-motor features of Parkinson’s disease. Nat Rev Neurosci. 2017;18:435–50.PubMedCrossRef
136.
Zurück zum Zitat Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–40.PubMedCrossRef Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–40.PubMedCrossRef
137.
Zurück zum Zitat Poewe W. Non-motor symptoms in Parkinson’s disease. Eur J Neurol. 2008;15(Suppl 1):14–20.PubMedCrossRef Poewe W. Non-motor symptoms in Parkinson’s disease. Eur J Neurol. 2008;15(Suppl 1):14–20.PubMedCrossRef
138.
Zurück zum Zitat Schapira AH, Tolosa E. Molecular and clinical prodrome of Parkinson’s disease: implications for treatment. Nat Rev Neurol. 2010;6:309–17.PubMedCrossRef Schapira AH, Tolosa E. Molecular and clinical prodrome of Parkinson’s disease: implications for treatment. Nat Rev Neurol. 2010;6:309–17.PubMedCrossRef
139.
Zurück zum Zitat Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease—a multicenter study. Park Relat Disord. 2009;15:490–4.CrossRef Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease—a multicenter study. Park Relat Disord. 2009;15:490–4.CrossRef
140.
Zurück zum Zitat Silveira-Moriyama L, Guedes LC, Kngsbury A, Ayling H, Shaw K, et al. Hyposmia in G2019S LRRK2-related parkinsonism: clinical and pathologic data. Neurology. 2008;71:1021–6.PubMedCrossRef Silveira-Moriyama L, Guedes LC, Kngsbury A, Ayling H, Shaw K, et al. Hyposmia in G2019S LRRK2-related parkinsonism: clinical and pathologic data. Neurology. 2008;71:1021–6.PubMedCrossRef
141.
142.
Zurück zum Zitat Vilas D, Ispierto L, Alvarez R, et al. Clinical and imaging markers in premotor LRRK2 G2019S mutation carriers. Parkinsonism Relat Disord. 2015;21:1170–6.PubMedCrossRef Vilas D, Ispierto L, Alvarez R, et al. Clinical and imaging markers in premotor LRRK2 G2019S mutation carriers. Parkinsonism Relat Disord. 2015;21:1170–6.PubMedCrossRef
143.
Zurück zum Zitat Ponsen MM, Stiffers D, Booij J, Van Eck-Smit BLF, Wolters EC, Berendse HW. Idiopathic hyposmia as a preclinical sign of Parkinson’s disease. Ann Neurol. 2004;56:173–81.PubMedCrossRef Ponsen MM, Stiffers D, Booij J, Van Eck-Smit BLF, Wolters EC, Berendse HW. Idiopathic hyposmia as a preclinical sign of Parkinson’s disease. Ann Neurol. 2004;56:173–81.PubMedCrossRef
144.
145.
Zurück zum Zitat Haugen J, Müller ML, Kotagal V, Albin RL, Koeppe RA, Scott PJ, et al. Prevalence of impaired odor identification in Parkinson disease with imaging evidence of nigrostriatal denervation. J Neural Transm. 2016;123:421–4.PubMedCrossRef Haugen J, Müller ML, Kotagal V, Albin RL, Koeppe RA, Scott PJ, et al. Prevalence of impaired odor identification in Parkinson disease with imaging evidence of nigrostriatal denervation. J Neural Transm. 2016;123:421–4.PubMedCrossRef
146.
Zurück zum Zitat Morley JF, Duda JE. Use of hyposmia and other non-motor symptoms to distinguish between drug-induced parkinsonism and Parkinson’s disease. J Parkinson Dis. 2014;4:169–73. Morley JF, Duda JE. Use of hyposmia and other non-motor symptoms to distinguish between drug-induced parkinsonism and Parkinson’s disease. J Parkinson Dis. 2014;4:169–73.
147.
Zurück zum Zitat Mesholam RI, Moberg PJ, Mahr RN, Doty RL. Olfaction in neurodegenerative disease: a meta-analysis of olfactory functioning in Alzheimer’s and Parkinson’s disease. Arch Neurol. 1998;55:84–90.PubMedCrossRef Mesholam RI, Moberg PJ, Mahr RN, Doty RL. Olfaction in neurodegenerative disease: a meta-analysis of olfactory functioning in Alzheimer’s and Parkinson’s disease. Arch Neurol. 1998;55:84–90.PubMedCrossRef
148.
Zurück zum Zitat Rahayel S, Frasnelli J, Joubert S. The effect of Alzheimer’s disease and Parkinson’s disease on olfaction: a meta-analysis. Behav Brain Res. 2012;231:60–74.PubMedCrossRef Rahayel S, Frasnelli J, Joubert S. The effect of Alzheimer’s disease and Parkinson’s disease on olfaction: a meta-analysis. Behav Brain Res. 2012;231:60–74.PubMedCrossRef
149.
Zurück zum Zitat Hawkes CH, Shephard BC. Selective anosmia in Parkinson’s disease? Lancet. 1993;341:435–6.PubMedCrossRef Hawkes CH, Shephard BC. Selective anosmia in Parkinson’s disease? Lancet. 1993;341:435–6.PubMedCrossRef
150.
Zurück zum Zitat Haehner A, Maboshe W, Baptista RB, Storch A, Reichmann H, Hummel T. Selective hyposmia in Parkinson’s disease? J Neurol. 2013;260:3158–60.CrossRef Haehner A, Maboshe W, Baptista RB, Storch A, Reichmann H, Hummel T. Selective hyposmia in Parkinson’s disease? J Neurol. 2013;260:3158–60.CrossRef
152.
Zurück zum Zitat Thacker EL, O’Reilly EJ, Weisskopf MG, et al. Temporal relationship between cigarette smoking and risk of Parkinson’s disease. Neurology. 2007;68:764–8.PubMedPubMedCentralCrossRef Thacker EL, O’Reilly EJ, Weisskopf MG, et al. Temporal relationship between cigarette smoking and risk of Parkinson’s disease. Neurology. 2007;68:764–8.PubMedPubMedCentralCrossRef
153.
Zurück zum Zitat Sharer JD, Leon-Sarmietno FE, Morley JF, Weintraub D, Doty RL. Olfactory dysfunction in Parkinson’s disease: positive effect of cigarette smoking. Mov Disord. 2015;30:859–62.PubMedCrossRef Sharer JD, Leon-Sarmietno FE, Morley JF, Weintraub D, Doty RL. Olfactory dysfunction in Parkinson’s disease: positive effect of cigarette smoking. Mov Disord. 2015;30:859–62.PubMedCrossRef
154.
Zurück zum Zitat Lucassen EB, Sterling NW, Lee EY, Cehn H, Lewis MM, Kong L, et al. History of smoking and olfaction in Parkinson’s disease. Mov Disord. 2014;29:1069–74.PubMedPubMedCentralCrossRef Lucassen EB, Sterling NW, Lee EY, Cehn H, Lewis MM, Kong L, et al. History of smoking and olfaction in Parkinson’s disease. Mov Disord. 2014;29:1069–74.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Witt M, Bormann K, Gudziol V, et al. Biopsies of olfactory epithelium in patients with Parkinson’s disease. Mov Disord. 2009;24:906–14.PubMedCrossRef Witt M, Bormann K, Gudziol V, et al. Biopsies of olfactory epithelium in patients with Parkinson’s disease. Mov Disord. 2009;24:906–14.PubMedCrossRef
156.
Zurück zum Zitat Hummel T, Witt M, Reichmnn H, Welge-Luessen A, Haehner A. Immunohistochemical, volumetric, and functional neuroimaging studies in patients with idiopathic Parkinson’s disease. J Neurol Sci. 2010;289:119–22.PubMedCrossRef Hummel T, Witt M, Reichmnn H, Welge-Luessen A, Haehner A. Immunohistochemical, volumetric, and functional neuroimaging studies in patients with idiopathic Parkinson’s disease. J Neurol Sci. 2010;289:119–22.PubMedCrossRef
157.
Zurück zum Zitat Schneider SA, Boettner M, Alexoudi A, Zorenkov D, Deuschl G, Wedel T. Can we use peripheral tissue biopsies to diagnose Parkinson’s disease? A review of the literature. Eur J Neurol. 2016;23:247–61.PubMedCrossRef Schneider SA, Boettner M, Alexoudi A, Zorenkov D, Deuschl G, Wedel T. Can we use peripheral tissue biopsies to diagnose Parkinson’s disease? A review of the literature. Eur J Neurol. 2016;23:247–61.PubMedCrossRef
158.
Zurück zum Zitat Brodoehl S, Klingner C, Volk GF, Bitter T, Witte OW, Redecker C. Decreased olfactory bulb volume in idiopathic Parkinson’s disease detected by 3.0-Tesla magnetic resonance imaging. Mov Disord. 2012;27:1019–25.PubMedCrossRef Brodoehl S, Klingner C, Volk GF, Bitter T, Witte OW, Redecker C. Decreased olfactory bulb volume in idiopathic Parkinson’s disease detected by 3.0-Tesla magnetic resonance imaging. Mov Disord. 2012;27:1019–25.PubMedCrossRef
159.
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;83:564–70.PubMedCrossRef Chen S, Tan HY, Wu ZH, Sun CP, He JX, Li XC, et al. Imaging of olfactory bulb and gray matter volumes in brain areas associated with olfactory function in patients with Parkinson’s disease and multiple system atrophy. Eur J Radiol. 2014;83:564–70.PubMedCrossRef
160.
Zurück zum Zitat Li J, Gu CZ, Su JB, Zhu LH, Zhou Y, Huang HY, et al. Changes in olfactory bulb volume in Parkinson’s disease: a systematic review and meta-analysis. PLoS One. 2016;11:e0149286.PubMedPubMedCentralCrossRef Li J, Gu CZ, Su JB, Zhu LH, Zhou Y, Huang HY, et al. Changes in olfactory bulb volume in Parkinson’s disease: a systematic review and meta-analysis. PLoS One. 2016;11:e0149286.PubMedPubMedCentralCrossRef
161.
Zurück zum Zitat Tanik N, Serin HI, Celikbilek A, Inan LE, Gundogdu F. Associations of olfactory bulb and depth of olfactory sulcus with basal ganglia and hippocampus in patients with Parkinson’s disease. Neurosci Lett. 2016;620:111–4.PubMedCrossRef Tanik N, Serin HI, Celikbilek A, Inan LE, Gundogdu F. Associations of olfactory bulb and depth of olfactory sulcus with basal ganglia and hippocampus in patients with Parkinson’s disease. Neurosci Lett. 2016;620:111–4.PubMedCrossRef
162.
Zurück zum Zitat Sengoku R, Matsushima S, Bono K, et al. Olfactory function combined with morphology distinguishes Parkinson’s disease. Parkinsonism Relat Disord. 2015;21:771–7.PubMedCrossRef Sengoku R, Matsushima S, Bono K, et al. Olfactory function combined with morphology distinguishes Parkinson’s disease. Parkinsonism Relat Disord. 2015;21:771–7.PubMedCrossRef
163.
Zurück zum Zitat Mueller A, Abolmaali ND, Hakimi AR, Gloeckler T, Herting B, Reichmann H, et al. Olfactory bulb volumes in patients with idiopathic Parkinson’s disease a pilot study. J Neural Transm. 2005;112:1363–70.PubMedCrossRef Mueller A, Abolmaali ND, Hakimi AR, Gloeckler T, Herting B, Reichmann H, et al. Olfactory bulb volumes in patients with idiopathic Parkinson’s disease a pilot study. J Neural Transm. 2005;112:1363–70.PubMedCrossRef
164.
Zurück zum Zitat Hakyemez HA, Veyseller B, Ozer F, Pzben S, Bayraktar GI, Gurbuz D, et al. Relationship of olfactory function with olfactory bulbs volume, disease duration and Unified Parkinson’s disease rating scale scores in patients with early stage of idiopathic Parkinson’s disease. J Clin Neurosci. 2013;20:1469–70.PubMedCrossRef Hakyemez HA, Veyseller B, Ozer F, Pzben S, Bayraktar GI, Gurbuz D, et al. Relationship of olfactory function with olfactory bulbs volume, disease duration and Unified Parkinson’s disease rating scale scores in patients with early stage of idiopathic Parkinson’s disease. J Clin Neurosci. 2013;20:1469–70.PubMedCrossRef
165.
Zurück zum Zitat Altinayar S, Oner S, Can S, Kizilay A, Kamisili S, Sarac K. Olfactory dysfunction and its relation olfactory bulb volume in Parkinson’s disease. Eur Rev Med Pharmacol Sci. 2014;18:3659–64.PubMed Altinayar S, Oner S, Can S, Kizilay A, Kamisili S, Sarac K. Olfactory dysfunction and its relation olfactory bulb volume in Parkinson’s disease. Eur Rev Med Pharmacol Sci. 2014;18:3659–64.PubMed
166.
Zurück zum Zitat Scherfler C, Schocke MF, Seppi K, Esterhammer R, Brenneis C, Jaschke W, et al. Voxel-wise analysis of diffusion weighted imaging reveals disruption of the olfactory tract in Parkinson’s disease. Brain. 2006;129:538–42.PubMedCrossRef Scherfler C, Schocke MF, Seppi K, Esterhammer R, Brenneis C, Jaschke W, et al. Voxel-wise analysis of diffusion weighted imaging reveals disruption of the olfactory tract in Parkinson’s disease. Brain. 2006;129:538–42.PubMedCrossRef
167.
Zurück zum Zitat Ibarretxe-Bilbao N, Junque C, Marti MJ, Valldeoriola F, Vendrell P, Bargallo N, et al. Olfactory impairment in Parkinson’s disease and white matter abnormalities in central olfactory areas: a voxel-based diffusion tensor imaging study. Mov Disord. 2010;25:1888–94.PubMedCrossRef Ibarretxe-Bilbao N, Junque C, Marti MJ, Valldeoriola F, Vendrell P, Bargallo N, et al. Olfactory impairment in Parkinson’s disease and white matter abnormalities in central olfactory areas: a voxel-based diffusion tensor imaging study. Mov Disord. 2010;25:1888–94.PubMedCrossRef
168.
Zurück zum Zitat Iannilli E, Stephan L, Hummel T, Reichmann H, Haehner A. Olfactory impairment in Parkinson’s disease is a consequence of central nervous system decline. J Neurol. 2017;264:1236–46.PubMedCrossRef Iannilli E, Stephan L, Hummel T, Reichmann H, Haehner A. Olfactory impairment in Parkinson’s disease is a consequence of central nervous system decline. J Neurol. 2017;264:1236–46.PubMedCrossRef
169.
Zurück zum Zitat Westermann B, Wattendorf E, Schwedtfeger U, et al. Functional imaging of the cerebral olfactory system in patients with Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2008;79:19–24.PubMedCrossRef Westermann B, Wattendorf E, Schwedtfeger U, et al. Functional imaging of the cerebral olfactory system in patients with Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2008;79:19–24.PubMedCrossRef
170.
Zurück zum Zitat Su M, Wang S, Fang W, Zhu Y, Li R, Sheng K, et al. Alterations in the limbic/paralimbic cirtices of Parkinson’s disease patients with hyposmia under resting-state functional MRI by regional homogeneity and functional connectivity analysis. Parkinsonism Relat Disord. 2015;21:698–703.PubMedCrossRef Su M, Wang S, Fang W, Zhu Y, Li R, Sheng K, et al. Alterations in the limbic/paralimbic cirtices of Parkinson’s disease patients with hyposmia under resting-state functional MRI by regional homogeneity and functional connectivity analysis. Parkinsonism Relat Disord. 2015;21:698–703.PubMedCrossRef
171.
Zurück zum Zitat Tolosa E, Gaig C, Santamaria J, Compta Y. Diagnosis and the premotor phase of Parkinson disease. Neurology. 2009;72(Suppl):S12–20.PubMedCrossRef Tolosa E, Gaig C, Santamaria J, Compta Y. Diagnosis and the premotor phase of Parkinson disease. Neurology. 2009;72(Suppl):S12–20.PubMedCrossRef
173.
Zurück zum Zitat Ross GW, Petrovich H, Abbot RD, et al. Association of olfactory dysfunction with risk for future Parkinsons’ disease. Ann Neurol. 2008;63:167–73.PubMedCrossRef Ross GW, Petrovich H, Abbot RD, et al. Association of olfactory dysfunction with risk for future Parkinsons’ disease. Ann Neurol. 2008;63:167–73.PubMedCrossRef
174.
Zurück zum Zitat Berendse HW, Booij J, Francot CM, Bergmans PL, Hijman R, Stoof JC, et al. Subclinical dopaminergic dysfunction in asymptomatic Parkinson’s disease patients’ relatives with a decreased sense of smell. Ann Neurol. 2001;50:34–41.PubMedCrossRef Berendse HW, Booij J, Francot CM, Bergmans PL, Hijman R, Stoof JC, et al. Subclinical dopaminergic dysfunction in asymptomatic Parkinson’s disease patients’ relatives with a decreased sense of smell. Ann Neurol. 2001;50:34–41.PubMedCrossRef
175.
Zurück zum Zitat Pont-Sunyer C, Hotter A, Haig C, et al. The onset of nomotor symptoms in Parkinson’s disease (the ONSET PD study). Mov Disord. 2015;30:229–37.PubMedCrossRef Pont-Sunyer C, Hotter A, Haig C, et al. The onset of nomotor symptoms in Parkinson’s disease (the ONSET PD study). Mov Disord. 2015;30:229–37.PubMedCrossRef
176.
Zurück zum Zitat Haehner A, Hummel T, Hummel C, Sommer U, Junghanns S, Reichmann H. Olfactory loss may be a first sign of idiopathic Parkinson’s disease. Mov Disord. 2007;22:839–42.PubMedCrossRef Haehner A, Hummel T, Hummel C, Sommer U, Junghanns S, Reichmann H. Olfactory loss may be a first sign of idiopathic Parkinson’s disease. Mov Disord. 2007;22:839–42.PubMedCrossRef
177.
Zurück zum Zitat Dickson DW, Fujishiro H, DellDonne A, et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 2008;115:437–44.PubMedCrossRef Dickson DW, Fujishiro H, DellDonne A, et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol. 2008;115:437–44.PubMedCrossRef
178.
Zurück zum Zitat Haehner A, Schöpf V, Loureiro A, Linn J, Reichmann H, Hummel T, et al. Susbtantia nigra fractional anisotropy changes confirm the PD at-risk status of paitients with idiopathic smell loss. Parkinsonism Relat Disord. 2018;50:113–6.PubMedCrossRef Haehner A, Schöpf V, Loureiro A, Linn J, Reichmann H, Hummel T, et al. Susbtantia nigra fractional anisotropy changes confirm the PD at-risk status of paitients with idiopathic smell loss. Parkinsonism Relat Disord. 2018;50:113–6.PubMedCrossRef
179.
Zurück zum Zitat Berendse HW, Ponsen MM. Diagnosing premotor Parkinson’s disease using a two-step approach combining olfactory testing and DAT SPECT imaging. Parkinsonism Relat Disord. 2009;15(Suppl 3):S26–30.PubMedCrossRef Berendse HW, Ponsen MM. Diagnosing premotor Parkinson’s disease using a two-step approach combining olfactory testing and DAT SPECT imaging. Parkinsonism Relat Disord. 2009;15(Suppl 3):S26–30.PubMedCrossRef
180.
Zurück zum Zitat Ponsen MM, Stoffers D, Wolters EC, Booij J, Berendse HW. Olfactory testing combined with dopamine transporter imaging as a method to detect prodromal Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81:396–9.PubMedCrossRef Ponsen MM, Stoffers D, Wolters EC, Booij J, Berendse HW. Olfactory testing combined with dopamine transporter imaging as a method to detect prodromal Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81:396–9.PubMedCrossRef
181.
Zurück zum Zitat Sierra M, Sánchez-Juan P, Martínez-Rodríguez MI, et al. Olfaction and imaging biomarkers in premotor LRRK2 G2019S-associated Parkinson disease. Neurology. 2013;80:621–6.PubMedCrossRef Sierra M, Sánchez-Juan P, Martínez-Rodríguez MI, et al. Olfaction and imaging biomarkers in premotor LRRK2 G2019S-associated Parkinson disease. Neurology. 2013;80:621–6.PubMedCrossRef
182.
Zurück zum Zitat Jennings D, Siderowf A, Stern M, Seibyil J, Eberly S, Oakes D, et al. Conversion to Parkinson Disease in the PARS hyposmic and dopamine transporter-deficit prodromal cohort. JAMA Neurol. 2017;74:933–40.PubMedCrossRefPubMedCentral Jennings D, Siderowf A, Stern M, Seibyil J, Eberly S, Oakes D, et al. Conversion to Parkinson Disease in the PARS hyposmic and dopamine transporter-deficit prodromal cohort. JAMA Neurol. 2017;74:933–40.PubMedCrossRefPubMedCentral
183.
Zurück zum Zitat Doty RL, Stern MB, Pfeiffer C, Gollomp SM, Hurtig HI. Bilateral olfactory dysfunction in early stage treated and untreated idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1992;55:138–42.PubMedPubMedCentralCrossRef Doty RL, Stern MB, Pfeiffer C, Gollomp SM, Hurtig HI. Bilateral olfactory dysfunction in early stage treated and untreated idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1992;55:138–42.PubMedPubMedCentralCrossRef
184.
Zurück zum Zitat Herting B, Schulze S, Reichmann H, Haehner A, Hummel T. A longitudinal study of olfactory function in patients with idiopathic Parkinson’s disease. J Neurol. 2008;255:367–70.PubMedCrossRef Herting B, Schulze S, Reichmann H, Haehner A, Hummel T. A longitudinal study of olfactory function in patients with idiopathic Parkinson’s disease. J Neurol. 2008;255:367–70.PubMedCrossRef
185.
Zurück zum Zitat Cavaco S, Gonçalves A, Mendes A, et al. Abnormal olfaction in Parkinson’s disease is related to faster disease progression. Behav Neurol. 2015: 976589. Cavaco S, Gonçalves A, Mendes A, et al. Abnormal olfaction in Parkinson’s disease is related to faster disease progression. Behav Neurol. 2015: 976589.
186.
Zurück zum Zitat Tissingh G, Berendse HW, Bergmans P, DeWaard R, Drukarch B, Stoof JC, et al. Loss of olfaction in de novo and treated Parkinson’s disease: possible implications for early diagnosis. Mov Disord. 2001;16:41–6.PubMedCrossRef Tissingh G, Berendse HW, Bergmans P, DeWaard R, Drukarch B, Stoof JC, et al. Loss of olfaction in de novo and treated Parkinson’s disease: possible implications for early diagnosis. Mov Disord. 2001;16:41–6.PubMedCrossRef
187.
Zurück zum Zitat Boesveldt S, Verbaan D, Knol DL, Visser M, van Rooden SM, van Hilten JJ, et al. A comparative study of odor identification and odor discrimination deficits in Parkinson’s disease. Mov Disord. 2008;23:1984–90.PubMedCrossRef Boesveldt S, Verbaan D, Knol DL, Visser M, van Rooden SM, van Hilten JJ, et al. A comparative study of odor identification and odor discrimination deficits in Parkinson’s disease. Mov Disord. 2008;23:1984–90.PubMedCrossRef
188.
Zurück zum Zitat Siderowf A, Newberg A, Chou KL, et al. [99mTc]TRODAT-1-SPECT imaging correlates with odor identification in early Parkinson disease. Neurology. 2005;64:1716–20.PubMedCrossRef Siderowf A, Newberg A, Chou KL, et al. [99mTc]TRODAT-1-SPECT imaging correlates with odor identification in early Parkinson disease. Neurology. 2005;64:1716–20.PubMedCrossRef
189.
Zurück zum Zitat Lee EY, Eslinger PJ, Du G, Kong L, Lewis MM, Huang X. Olfactory-related cortical atrophy is associated with olfactory dysfunction in Parkinson’s disease. Mov Disord. 2014;29:1205–8.PubMedPubMedCentralCrossRef Lee EY, Eslinger PJ, Du G, Kong L, Lewis MM, Huang X. Olfactory-related cortical atrophy is associated with olfactory dysfunction in Parkinson’s disease. Mov Disord. 2014;29:1205–8.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Fullard ME, Tran B, Xie SX, et al. Olfactory impairment predicts cognitive decline in early Parkinson’s disease. Parkinsonism Relat Disord. 2016;25:45–51.PubMedPubMedCentralCrossRef Fullard ME, Tran B, Xie SX, et al. Olfactory impairment predicts cognitive decline in early Parkinson’s disease. Parkinsonism Relat Disord. 2016;25:45–51.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Kang SH, Lee HM, Seo WK, Kim JH, Koh SB. The combined effect of REM sleep behavior disorder and hyposmia on cognition and motor phenotype in Parkinson’s disease. J Neurol Sci. 2016;368:374–8.PubMedCrossRef Kang SH, Lee HM, Seo WK, Kim JH, Koh SB. The combined effect of REM sleep behavior disorder and hyposmia on cognition and motor phenotype in Parkinson’s disease. J Neurol Sci. 2016;368:374–8.PubMedCrossRef
192.
Zurück zum Zitat Domellöf ME, Lundin KF, Edström M, Forsgren L. Olfactory dysfunction and dementia in newly diagnosed patients with Parkinson’s disease. Parkinsonism Relat Disord. 2017;38:41–7.PubMedCrossRef Domellöf ME, Lundin KF, Edström M, Forsgren L. Olfactory dysfunction and dementia in newly diagnosed patients with Parkinson’s disease. Parkinsonism Relat Disord. 2017;38:41–7.PubMedCrossRef
193.
Zurück zum Zitat Stephenson R, Houghton D, Sundarararjan S, Doty RL, Stern M, Xie SX, et al. Odor identification deficits are associated with increased risk of neuropsychiatric complications in patients with Parkinson’s disease. Mov Disord. 2010;25:2099–104.PubMedPubMedCentralCrossRef Stephenson R, Houghton D, Sundarararjan S, Doty RL, Stern M, Xie SX, et al. Odor identification deficits are associated with increased risk of neuropsychiatric complications in patients with Parkinson’s disease. Mov Disord. 2010;25:2099–104.PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat Baba T, Kikuchi A, Hirayama K, et al. Severe olfactory dysfunction is a prodromal symptom or dementia associated with Parkinson’s disease: a 3 years longitudinal study. Brain. 2012;135:161–9.PubMedCrossRef Baba T, Kikuchi A, Hirayama K, et al. Severe olfactory dysfunction is a prodromal symptom or dementia associated with Parkinson’s disease: a 3 years longitudinal study. Brain. 2012;135:161–9.PubMedCrossRef
196.
Zurück zum Zitat Bibi M, Mollenhauer B, Esselmann H, et al. CSF amyloid-beta-peptides in Alzheimer’s disease dementia, dementia with Lewy bodies and Parkinson’s disease dementia. Brain. 2006;129:1177–87.CrossRef Bibi M, Mollenhauer B, Esselmann H, et al. CSF amyloid-beta-peptides in Alzheimer’s disease dementia, dementia with Lewy bodies and Parkinson’s disease dementia. Brain. 2006;129:1177–87.CrossRef
197.
Zurück zum Zitat Compta Y, Santamaría J, Ratti L, Tolosa E, Iranzo A, Muñoz E, et al. Cerebrospinal hypocretin, daytime sleepiness and sleep architecture in Parkinson’s dementia. Brain. 2009;132:3308–17.PubMedCrossRef Compta Y, Santamaría J, Ratti L, Tolosa E, Iranzo A, Muñoz E, et al. Cerebrospinal hypocretin, daytime sleepiness and sleep architecture in Parkinson’s dementia. Brain. 2009;132:3308–17.PubMedCrossRef
198.
199.
Zurück zum Zitat Parnetti L, Farotti L, Eusebi P, et al. Differential role of CSF alpha-synuclein species, tau, and Aβ42 in Parkinson’s disease. Front Aging Neurosci. 2014;6:53.PubMedPubMedCentralCrossRef Parnetti L, Farotti L, Eusebi P, et al. Differential role of CSF alpha-synuclein species, tau, and Aβ42 in Parkinson’s disease. Front Aging Neurosci. 2014;6:53.PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Goldstein DS, Holmes C, Bentho O, et al. Biomarkers to detect central dopamine deficiency and distinguish Parkinson disease from multiple atrophy. Parkinsonism Relat Disord. 2008;14:600–7.PubMedPubMedCentralCrossRef Goldstein DS, Holmes C, Bentho O, et al. Biomarkers to detect central dopamine deficiency and distinguish Parkinson disease from multiple atrophy. Parkinsonism Relat Disord. 2008;14:600–7.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Silveira-Moriyama L, Hughes G, Church A, et al. Hyposmia in progressive supranuclear palsy. Mov Disord. 2010;25:570–7.PubMedCrossRef Silveira-Moriyama L, Hughes G, Church A, et al. Hyposmia in progressive supranuclear palsy. Mov Disord. 2010;25:570–7.PubMedCrossRef
202.
Zurück zum Zitat Suzuki M, Hashimoto M, Yoshioka M, Murakami M, Kawasaki K, Urashima M. The odor stick identification test for Japanese differentiates Parkinson’s disease form multiple system atrophy and progressive supranuclear palsy. BMC Neurol. 2011;11:157.PubMedPubMedCentralCrossRef Suzuki M, Hashimoto M, Yoshioka M, Murakami M, Kawasaki K, Urashima M. The odor stick identification test for Japanese differentiates Parkinson’s disease form multiple system atrophy and progressive supranuclear palsy. BMC Neurol. 2011;11:157.PubMedPubMedCentralCrossRef
203.
Zurück zum Zitat Glass PG, Lees AJ, Mathias C, et al. Olfaction in pathologically proven patients with multiple system atrophy. Mov Disord. 2012;27:327–8.PubMedCrossRef Glass PG, Lees AJ, Mathias C, et al. Olfaction in pathologically proven patients with multiple system atrophy. Mov Disord. 2012;27:327–8.PubMedCrossRef
204.
Zurück zum Zitat Wenning GK, Shephard B, hawkes C, Petruckevitch A, Lees A, Quinn N. Olfactory function in atypical parkinsonian syndromes. Acta Neurol Scand. 1995;81:247–50.CrossRef Wenning GK, Shephard B, hawkes C, Petruckevitch A, Lees A, Quinn N. Olfactory function in atypical parkinsonian syndromes. Acta Neurol Scand. 1995;81:247–50.CrossRef
205.
Zurück zum Zitat Müller A, Müngersdorf M, Reichmann H, Strehle G, Hummel T. Olfactory function in parkinsonian syndromes. J Clin Neurosci. 2002;9:521–4.PubMedCrossRef Müller A, Müngersdorf M, Reichmann H, Strehle G, Hummel T. Olfactory function in parkinsonian syndromes. J Clin Neurosci. 2002;9:521–4.PubMedCrossRef
206.
Zurück zum Zitat Brigo F, Erro R, Marangi A, Bhatia K, Tinazzi M. Differentiating drug-induced parkinsonism from Parkinson’s disease: an update on non-motor symptoms and investigations. Parkinsonism Relat Disord. 2014;20:808–14.PubMedCrossRef Brigo F, Erro R, Marangi A, Bhatia K, Tinazzi M. Differentiating drug-induced parkinsonism from Parkinson’s disease: an update on non-motor symptoms and investigations. Parkinsonism Relat Disord. 2014;20:808–14.PubMedCrossRef
207.
Zurück zum Zitat Katzenschlager R, Lees AJ. Olfaction and Parkinson’s syndromes: its role in differential diagnosis. Curr opin Neurol. 2004;17:417–23.PubMedCrossRef Katzenschlager R, Lees AJ. Olfaction and Parkinson’s syndromes: its role in differential diagnosis. Curr opin Neurol. 2004;17:417–23.PubMedCrossRef
208.
Zurück zum Zitat Kertelge L, Brüggemann N, Schmidt A, et al. Impaired sense of smell and color discrimination in monogenic and idiopathic Parkinson’s disease. Mov Disord. 2010;25:2665–9.PubMedCrossRef Kertelge L, Brüggemann N, Schmidt A, et al. Impaired sense of smell and color discrimination in monogenic and idiopathic Parkinson’s disease. Mov Disord. 2010;25:2665–9.PubMedCrossRef
209.
Zurück zum Zitat Paisán-Ruiz C, Lewis PA, Singleton AB. LRRK2: cause, risk, and mechanism. J Parkinson Dis. 2013;3:85–103. Paisán-Ruiz C, Lewis PA, Singleton AB. LRRK2: cause, risk, and mechanism. J Parkinson Dis. 2013;3:85–103.
210.
Zurück zum Zitat Silveira-Moriyama L, Munhoz RP, de J Carvalho M, et al. Olfactory heterogeneity in LRRK2 related parkinsonism. Mov Disord. 2010;25:2879–83.PubMedCrossRef Silveira-Moriyama L, Munhoz RP, de J Carvalho M, et al. Olfactory heterogeneity in LRRK2 related parkinsonism. Mov Disord. 2010;25:2879–83.PubMedCrossRef
211.
Zurück zum Zitat Bardien S, Lesage S, Brice A, Carr J. Genetic characteristic of leucine-rich repeat kinase 2 (LRRK2) associated Parkinson’s disease. Parkinsonism Relat Disord. 2011;17:501–8.PubMedCrossRef Bardien S, Lesage S, Brice A, Carr J. Genetic characteristic of leucine-rich repeat kinase 2 (LRRK2) associated Parkinson’s disease. Parkinsonism Relat Disord. 2011;17:501–8.PubMedCrossRef
212.
213.
Zurück zum Zitat Johansen KK, Waro BJ, Aasly JO. Olfactory dysfunction in sporadic Parkinson’s disease and LRRK2 carriers. Acta Neurol Scand. 2014;129:300–6.PubMedCrossRef Johansen KK, Waro BJ, Aasly JO. Olfactory dysfunction in sporadic Parkinson’s disease and LRRK2 carriers. Acta Neurol Scand. 2014;129:300–6.PubMedCrossRef
214.
Zurück zum Zitat Saunders-Pullman R, Mirelman A, Wang C, et al. Olfactory identification in LRRK2 G2019S mutation carriers: a relevant marker? Ann Clin Transl Neurol. 2014;1:670–8.PubMedPubMedCentralCrossRef Saunders-Pullman R, Mirelman A, Wang C, et al. Olfactory identification in LRRK2 G2019S mutation carriers: a relevant marker? Ann Clin Transl Neurol. 2014;1:670–8.PubMedPubMedCentralCrossRef
215.
Zurück zum Zitat Pont-Sunyer C, Tolosa E, Caspell-Garcia C, et al. The prodromal phase of leucine-rich repeat kinase 2-associated Parkinson’s disease: clinical and imaging studies. Mov Disord. 2017;32:726–38.PubMedCrossRef Pont-Sunyer C, Tolosa E, Caspell-Garcia C, et al. The prodromal phase of leucine-rich repeat kinase 2-associated Parkinson’s disease: clinical and imaging studies. Mov Disord. 2017;32:726–38.PubMedCrossRef
216.
Zurück zum Zitat Mirelman A, Heman T, Yasinovsky K, et al. Fall risk and gait in Parkinson’s disease: the role of the LRRK2 G2019S mutation. Mov Disord. 2013;28:1683–90.PubMedCrossRef Mirelman A, Heman T, Yasinovsky K, et al. Fall risk and gait in Parkinson’s disease: the role of the LRRK2 G2019S mutation. Mov Disord. 2013;28:1683–90.PubMedCrossRef
217.
Zurück zum Zitat Poulopulos M, Levy OA, Alcalay RN. The neuropathology of genetic Parkinson’s disease. Mov Disord. 2012;27:831–42.CrossRef Poulopulos M, Levy OA, Alcalay RN. The neuropathology of genetic Parkinson’s disease. Mov Disord. 2012;27:831–42.CrossRef
218.
Zurück zum Zitat Kalia LV, Lang AE, Hazrati LN, et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson’s disease. JAMA Neurol. 2015;72:100–5.PubMedPubMedCentralCrossRef Kalia LV, Lang AE, Hazrati LN, et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson’s disease. JAMA Neurol. 2015;72:100–5.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Sierra M, Martínez-Rodríguez I, Sánchez-Juan P, et al. Prospective clinical and DaT-SPECT imaging in premotor LRRK2 G2019S-associated Parkinson disease. Neurology. 2017;89:439–44.PubMedPubMedCentralCrossRef Sierra M, Martínez-Rodríguez I, Sánchez-Juan P, et al. Prospective clinical and DaT-SPECT imaging in premotor LRRK2 G2019S-associated Parkinson disease. Neurology. 2017;89:439–44.PubMedPubMedCentralCrossRef
220.
Zurück zum Zitat Bostantjopoulou S, Katsarou Z, Papadimitriou A, Veletza V, Hatzigeorgiou G, Lees A. Clinical features of parkinsonian patients with the alpha-synuclein (G209A) mutation. Mov Disord. 2001;16:1007–13.PubMedCrossRef Bostantjopoulou S, Katsarou Z, Papadimitriou A, Veletza V, Hatzigeorgiou G, Lees A. Clinical features of parkinsonian patients with the alpha-synuclein (G209A) mutation. Mov Disord. 2001;16:1007–13.PubMedCrossRef
221.
Zurück zum Zitat Tijero B, Gomez-Esteban JC, Llorens V, et al. Cardiac sympathetic denervation precedes nigrostriatal loss in the E46K mutation of the alpha-synuclein gene (SNCA). Clin Auton Res. 2010;20:267–9.PubMedCrossRef Tijero B, Gomez-Esteban JC, Llorens V, et al. Cardiac sympathetic denervation precedes nigrostriatal loss in the E46K mutation of the alpha-synuclein gene (SNCA). Clin Auton Res. 2010;20:267–9.PubMedCrossRef
222.
Zurück zum Zitat Papadimitriou D, Antonelou R, Miligkos M, et al. Motor and non-motor features of carriers of the p.A53T alpha-synuclein mutation: A longitudinal study. Mov Disord. 2016;31:1226–30.PubMedCrossRef Papadimitriou D, Antonelou R, Miligkos M, et al. Motor and non-motor features of carriers of the p.A53T alpha-synuclein mutation: A longitudinal study. Mov Disord. 2016;31:1226–30.PubMedCrossRef
223.
Zurück zum Zitat Alcalay RN, Siderowf A, Ottman R, et al. Olfaction in Parkin heterozygotes and compound heterozygotes: the CORE-PD study. Neurology. 2011;76:319–26.PubMedCrossRef Alcalay RN, Siderowf A, Ottman R, et al. Olfaction in Parkin heterozygotes and compound heterozygotes: the CORE-PD study. Neurology. 2011;76:319–26.PubMedCrossRef
224.
Zurück zum Zitat Malek N, Swallow DM, Grosset KA, et al. Olfaction in Parkin single and compound heterozygotes in a cohort of young onset Parkinson’s disease patients. Acta Neurol Scand. 2016;134:271–6.PubMedCrossRef Malek N, Swallow DM, Grosset KA, et al. Olfaction in Parkin single and compound heterozygotes in a cohort of young onset Parkinson’s disease patients. Acta Neurol Scand. 2016;134:271–6.PubMedCrossRef
225.
Zurück zum Zitat Ferraris A, Ialongo T, Passali GC, et al. Olfactory dysfunction in Parkinsonism caused by PINK1 mutations. Mov Disord. 2009;24:2350–7.PubMed Ferraris A, Ialongo T, Passali GC, et al. Olfactory dysfunction in Parkinsonism caused by PINK1 mutations. Mov Disord. 2009;24:2350–7.PubMed
226.
Zurück zum Zitat Saunders-Pullman R, Hagenah J, Dhawan V, et al. Gaucher disease ascertained through a Parkinson’s center: imaging and clinical characterization. Mov Disord. 2010;25:1364–72.PubMedPubMedCentralCrossRef Saunders-Pullman R, Hagenah J, Dhawan V, et al. Gaucher disease ascertained through a Parkinson’s center: imaging and clinical characterization. Mov Disord. 2010;25:1364–72.PubMedPubMedCentralCrossRef
227.
Zurück zum Zitat McNeill A, Duran R, Proukakis C, et al. Hyposmia and cognitive impairment in Gaucher disease patients and carriers. Mov Disord. 2012;27:526–32.PubMedPubMedCentralCrossRef McNeill A, Duran R, Proukakis C, et al. Hyposmia and cognitive impairment in Gaucher disease patients and carriers. Mov Disord. 2012;27:526–32.PubMedPubMedCentralCrossRef
228.
Zurück zum Zitat Beavan M, McNeill A, Proukakis CH, et al. Evolution of prodromal clinical markers of Parkinson disease in a glucocerebrosidase mutation positive cohort. JAMA Neurol. 2015;72:201–8.PubMedPubMedCentralCrossRef Beavan M, McNeill A, Proukakis CH, et al. Evolution of prodromal clinical markers of Parkinson disease in a glucocerebrosidase mutation positive cohort. JAMA Neurol. 2015;72:201–8.PubMedPubMedCentralCrossRef
229.
Zurück zum Zitat Del Tredici K, Rüb U, De Vos RA, Bohl JR, Braak H. Where does Parkinson disease pathology begin in the brain? J Neuropathol Exp Neurol. 2002;61:413–26.PubMedCrossRef Del Tredici K, Rüb U, De Vos RA, Bohl JR, Braak H. Where does Parkinson disease pathology begin in the brain? J Neuropathol Exp Neurol. 2002;61:413–26.PubMedCrossRef
230.
Zurück zum Zitat Silveira-Moriyama L, Holton JL, Kngsbury A, Ayling H, Petrie A, Sterlacci W, et al. Regional differences in the severity of Lewy body pathology across the olfactory cortex. Neurosci Lett. 2009;453:77–80.PubMedCrossRef Silveira-Moriyama L, Holton JL, Kngsbury A, Ayling H, Petrie A, Sterlacci W, et al. Regional differences in the severity of Lewy body pathology across the olfactory cortex. Neurosci Lett. 2009;453:77–80.PubMedCrossRef
231.
Zurück zum Zitat •• Rey NL, Steiner JA, Maroof N, Luk KC, Madaj Z, Trojanowski JQ, et al. Widespread transneuronal propagation of α-synucleopathy triggered in olfactory bulb mimics prodromal Parkinson’s disease. J Exp Med. 2016;213:1759–78. Preclinical evidence that α-synuclein fibrils injected at the olfactory bulb level induces olfactory deficits and spread to multiple brain regions, being the first evidence of a transneuronal, progressive propagation of Parkinson’s diasease (PD)-like α-synuclein pathology. PubMedPubMedCentralCrossRef •• Rey NL, Steiner JA, Maroof N, Luk KC, Madaj Z, Trojanowski JQ, et al. Widespread transneuronal propagation of α-synucleopathy triggered in olfactory bulb mimics prodromal Parkinson’s disease. J Exp Med. 2016;213:1759–78. Preclinical evidence that α-synuclein fibrils injected at the olfactory bulb level induces olfactory deficits and spread to multiple brain regions, being the first evidence of a transneuronal, progressive propagation of Parkinson’s diasease (PD)-like α-synuclein pathology. PubMedPubMedCentralCrossRef
232.
Zurück zum Zitat Mason DM, Nouraei N, Pant DB, Miner KM, Hutchinson DF, Luk KC, et al. Transmission of α-synucleinopathy from olfactory structures deep into the temporal lobe. Mol Neurodegener. 2016;11:49.PubMedPubMedCentralCrossRef Mason DM, Nouraei N, Pant DB, Miner KM, Hutchinson DF, Luk KC, et al. Transmission of α-synucleinopathy from olfactory structures deep into the temporal lobe. Mol Neurodegener. 2016;11:49.PubMedPubMedCentralCrossRef
233.
Zurück zum Zitat Duda JE, Shah E, Arnold SE, Lee VM, Tojanowski JQ. The expression of alpha-, beta-, and gamma-synucleins in olfactory mucosa from patients with and without neurodegenerative disease. Exp Neurol. 1999;160:515–22.PubMedCrossRef Duda JE, Shah E, Arnold SE, Lee VM, Tojanowski JQ. The expression of alpha-, beta-, and gamma-synucleins in olfactory mucosa from patients with and without neurodegenerative disease. Exp Neurol. 1999;160:515–22.PubMedCrossRef
234.
Zurück zum Zitat Ferrer I, López-González I, Carmona M, Dalfó E, Pujol A, Martínez A. Neurochemistry and the non-motor aspects of PD. Neurobiol Dis. 2012;46:508–26.PubMedCrossRef Ferrer I, López-González I, Carmona M, Dalfó E, Pujol A, Martínez A. Neurochemistry and the non-motor aspects of PD. Neurobiol Dis. 2012;46:508–26.PubMedCrossRef
235.
Zurück zum Zitat Huisman E, Uylings HB, Hoogland PV. A 100% increase of dopaminergic cells in the olfactory bulb may explain hyposmia in Parkinsons’s disease. Mov Disord. 2004;19:687–92.PubMedCrossRef Huisman E, Uylings HB, Hoogland PV. A 100% increase of dopaminergic cells in the olfactory bulb may explain hyposmia in Parkinsons’s disease. Mov Disord. 2004;19:687–92.PubMedCrossRef
236.
Zurück zum Zitat Huisman E, Uylings HB, Hoogland PV. Gender-related changes in increase of dopaminergic neurons in the olfactory bulb of Parkinson’s disease patients. Mov Disord. 2008;23:1407–13.PubMedCrossRef Huisman E, Uylings HB, Hoogland PV. Gender-related changes in increase of dopaminergic neurons in the olfactory bulb of Parkinson’s disease patients. Mov Disord. 2008;23:1407–13.PubMedCrossRef
237.
Zurück zum Zitat Mundiñano LC, Caballero MC, Ordóñez C, et al. Increased dopaminergic cells and protein aggregates in the olfactory bulb of patients with neurodegenerative disorders. Acta Neuropathol. 2011;122:61–74.PubMedCrossRef Mundiñano LC, Caballero MC, Ordóñez C, et al. Increased dopaminergic cells and protein aggregates in the olfactory bulb of patients with neurodegenerative disorders. Acta Neuropathol. 2011;122:61–74.PubMedCrossRef
238.
Zurück zum Zitat Hubbard PS, Esiri MM, Reading M, MCShane R, Nagy Z. Alpha-synuclein pathology in the olfactory pathways of dementia patients. J Anat. 2007;211:117–24.PubMedPubMedCentralCrossRef Hubbard PS, Esiri MM, Reading M, MCShane R, Nagy Z. Alpha-synuclein pathology in the olfactory pathways of dementia patients. J Anat. 2007;211:117–24.PubMedPubMedCentralCrossRef
239.
Zurück zum Zitat Ubeda-Bañón I, Flores-Cuadrado A, Saiz-Sanchez D, Martinez-Marcos A. Differential effects of Parkinson’s disease on interneuron subtypes within the human anterior olfactory nucleus. Front Neuroanat. 2017;11:113.PubMedPubMedCentralCrossRef Ubeda-Bañón I, Flores-Cuadrado A, Saiz-Sanchez D, Martinez-Marcos A. Differential effects of Parkinson’s disease on interneuron subtypes within the human anterior olfactory nucleus. Front Neuroanat. 2017;11:113.PubMedPubMedCentralCrossRef
240.
Zurück zum Zitat Harding AJ, Stimson E, Henderson JM, Halliday GM. Clinical correlates of selective pathology in the amygdala of patients with Parkinson’s disease. Brain. 2002;125:2431–45.PubMedCrossRef Harding AJ, Stimson E, Henderson JM, Halliday GM. Clinical correlates of selective pathology in the amygdala of patients with Parkinson’s disease. Brain. 2002;125:2431–45.PubMedCrossRef
241.
Zurück zum Zitat Wattendorf E, Welge-Lüssen A, Fiedler K, Bilecen D, Wolfensberger M, Fuhr P, et al. Olfactory impairment predicts brain atrophy in Parkinson’s disease. J Neurosci. 2009;29:15410–3.PubMedCrossRef Wattendorf E, Welge-Lüssen A, Fiedler K, Bilecen D, Wolfensberger M, Fuhr P, et al. Olfactory impairment predicts brain atrophy in Parkinson’s disease. J Neurosci. 2009;29:15410–3.PubMedCrossRef
242.
Zurück zum Zitat Tsuboi Y, Wszolek ZK, Graff-Radford NR, Cookson N, Dickson DW. Tau pathology in the olfactory bulb correlates with Braak stage, Lewy body pathology and apolipoprotein epsilon4. Neuropathol Appl Neurobiol. 2003;29:503–10.PubMedCrossRef Tsuboi Y, Wszolek ZK, Graff-Radford NR, Cookson N, Dickson DW. Tau pathology in the olfactory bulb correlates with Braak stage, Lewy body pathology and apolipoprotein epsilon4. Neuropathol Appl Neurobiol. 2003;29:503–10.PubMedCrossRef
244.
Zurück zum Zitat Levey AI, Hersch SM, Rye DB, Sunahara RK, Niznik HB, Kitt CA, et al. Localization of D1 and D2 dopamine receptors in brain with subtype-specific antibodies. Proc Natl Acada Sci USA. 1993;90:8861–5.CrossRef Levey AI, Hersch SM, Rye DB, Sunahara RK, Niznik HB, Kitt CA, et al. Localization of D1 and D2 dopamine receptors in brain with subtype-specific antibodies. Proc Natl Acada Sci USA. 1993;90:8861–5.CrossRef
245.
Zurück zum Zitat Gutierrez-Mecinas M, Crespo C, Blasco-Ibáñez JM, et al. Distribution of D2 dopamine receptor in the olfactory glomeruli of the rat olfactory bulb. Eur J Neurosci. 2005;22:1357–67.PubMedCrossRef Gutierrez-Mecinas M, Crespo C, Blasco-Ibáñez JM, et al. Distribution of D2 dopamine receptor in the olfactory glomeruli of the rat olfactory bulb. Eur J Neurosci. 2005;22:1357–67.PubMedCrossRef
246.
Zurück zum Zitat Doty RL, Risser JM. Influence of the D-2 dopamine receptor agonist quinpirole on the odor detection performance of rats before and after spiperone administration. Psychopharmacology. 1989;98:310–5.PubMedCrossRef Doty RL, Risser JM. Influence of the D-2 dopamine receptor agonist quinpirole on the odor detection performance of rats before and after spiperone administration. Psychopharmacology. 1989;98:310–5.PubMedCrossRef
247.
Zurück zum Zitat Escanilla O, Yuhas C, Marzan D, Linster C. Dopaminergic modulation of olfactory bulb processing affects odor discrimination learning in rats. Behav Neurosci. 2009;123:828–33.PubMedPubMedCentralCrossRef Escanilla O, Yuhas C, Marzan D, Linster C. Dopaminergic modulation of olfactory bulb processing affects odor discrimination learning in rats. Behav Neurosci. 2009;123:828–33.PubMedPubMedCentralCrossRef
248.
Zurück zum Zitat Wei CJ, Linster C, Cleland TA. Dopamine D2 receptor activation modulates perceived odor intensity. Behav Neurosci. 2006;120:393–400.PubMedCrossRef Wei CJ, Linster C, Cleland TA. Dopamine D2 receptor activation modulates perceived odor intensity. Behav Neurosci. 2006;120:393–400.PubMedCrossRef
249.
Zurück zum Zitat Bastien-Dionne PO, David LS, Parent A, Saghatelyan A. Role of sensory activity on chemospecific populations of interneurons in the adult olfactory bulb. J Comp Neurol. 2010;518:1847–61.PubMedCrossRef Bastien-Dionne PO, David LS, Parent A, Saghatelyan A. Role of sensory activity on chemospecific populations of interneurons in the adult olfactory bulb. J Comp Neurol. 2010;518:1847–61.PubMedCrossRef
250.
Zurück zum Zitat Sawada M, Kaneko N, Inada H, Wake H, Kato Y, Yanagawa Y, et al. Sensory input regulates spatial and subtype-specific patterns of neuronal turnover in the adult olfactory bulb. J Neurosci. 2011;31:11587–96.PubMedCrossRef Sawada M, Kaneko N, Inada H, Wake H, Kato Y, Yanagawa Y, et al. Sensory input regulates spatial and subtype-specific patterns of neuronal turnover in the adult olfactory bulb. J Neurosci. 2011;31:11587–96.PubMedCrossRef
251.
Zurück zum Zitat Motles E, Tetas M, Gomez A. Behavioral effects evoked by SKF38393 and LY171555 in adult cats. Physiol Behav. 1995;57:983–8.PubMedCrossRef Motles E, Tetas M, Gomez A. Behavioral effects evoked by SKF38393 and LY171555 in adult cats. Physiol Behav. 1995;57:983–8.PubMedCrossRef
252.
Zurück zum Zitat Deeb J, Shah M, Muhammed N, Gunasekera R, Gannon K, Findley LJ, et al. A basic smell test is as sensitive as a dopamine transporter scan: comparison of olfaction, taste and DaTSCAN in the diagnosis of Parkinson’s disease. QJM. 2010;103:941–52.PubMedCrossRef Deeb J, Shah M, Muhammed N, Gunasekera R, Gannon K, Findley LJ, et al. A basic smell test is as sensitive as a dopamine transporter scan: comparison of olfaction, taste and DaTSCAN in the diagnosis of Parkinson’s disease. QJM. 2010;103:941–52.PubMedCrossRef
253.
Zurück zum Zitat Bohnen NI, Gedela S, Kuwabara H, Constantine GM, Mathis CA, Studenski SA, et al. Selective hyposmia and nigrostriatal dopaminergic denervation in Parkinson’s disease. J Neurol. 2007;254:84–90.PubMedCrossRef Bohnen NI, Gedela S, Kuwabara H, Constantine GM, Mathis CA, Studenski SA, et al. Selective hyposmia and nigrostriatal dopaminergic denervation in Parkinson’s disease. J Neurol. 2007;254:84–90.PubMedCrossRef
254.
Zurück zum Zitat Winner B, Desplats P, Hagl C, et al. Dopamine receptor activation promotes adult neurogenesis in an acute Parkinson model. Exp Neurol. 2009;219:543–52.PubMedPubMedCentralCrossRef Winner B, Desplats P, Hagl C, et al. Dopamine receptor activation promotes adult neurogenesis in an acute Parkinson model. Exp Neurol. 2009;219:543–52.PubMedPubMedCentralCrossRef
255.
Zurück zum Zitat Zaborsky L, Carlsen J, Brashear HR, Heimer L. Cholinergic afferents to the olfactory bulb in the rat with special emphasis on the projection neurons in the nucleus of the horizontal limb of the diagonal band. J Comp Neurol. 1986;243:488–509.CrossRef Zaborsky L, Carlsen J, Brashear HR, Heimer L. Cholinergic afferents to the olfactory bulb in the rat with special emphasis on the projection neurons in the nucleus of the horizontal limb of the diagonal band. J Comp Neurol. 1986;243:488–509.CrossRef
256.
Zurück zum Zitat D’Souza RD, Vijayaraghavan S. Nicotinic receptor-mediated filtering of mitral cell responses to olfactory nerve inputs involves the a3b4 subtype. J Neurosci. 2012;32:3261–6.PubMedPubMedCentralCrossRef D’Souza RD, Vijayaraghavan S. Nicotinic receptor-mediated filtering of mitral cell responses to olfactory nerve inputs involves the a3b4 subtype. J Neurosci. 2012;32:3261–6.PubMedPubMedCentralCrossRef
257.
Zurück zum Zitat D’Souza RD, Parsa PC, Vijayaraghavan S. Nicotinic receptors modulate olfactory bulb external tufted cells via an excitation-dependent inhibitory mechanism. J Neurophsyiol. 2013;110:1544–53.CrossRef D’Souza RD, Parsa PC, Vijayaraghavan S. Nicotinic receptors modulate olfactory bulb external tufted cells via an excitation-dependent inhibitory mechanism. J Neurophsyiol. 2013;110:1544–53.CrossRef
258.
Zurück zum Zitat Li G, Linster C, Cleland TA. Functional differentiation of cholinergic and noradrenergic modulation in a biophysical model of olfactory bulb granule cells. J Neurophsyiol. 2015;114:3177–200.CrossRef Li G, Linster C, Cleland TA. Functional differentiation of cholinergic and noradrenergic modulation in a biophysical model of olfactory bulb granule cells. J Neurophsyiol. 2015;114:3177–200.CrossRef
259.
Zurück zum Zitat Doty RL, Bagla R, Kim N. Physostigmine enhances performance on an odor mixture discrimination test. Physiol Behav. 1999;65:801–4.PubMedCrossRef Doty RL, Bagla R, Kim N. Physostigmine enhances performance on an odor mixture discrimination test. Physiol Behav. 1999;65:801–4.PubMedCrossRef
260.
261.
Zurück zum Zitat Mandairon N, Peace ST, Boudadi K, Boxhorn CE, Narla VA, Suffis SD, et al. Compensatory responses to age-related decline in odor quality acuity: cholinergic neuromodulation and olfactory enrichment. Neurobiol Aging. 2011;32:2254–65.PubMedCrossRef Mandairon N, Peace ST, Boudadi K, Boxhorn CE, Narla VA, Suffis SD, et al. Compensatory responses to age-related decline in odor quality acuity: cholinergic neuromodulation and olfactory enrichment. Neurobiol Aging. 2011;32:2254–65.PubMedCrossRef
262.
Zurück zum Zitat Devore S, Manella LC, Linster C. Blocking muscarinic receptors in the olfactory bulb impairs performance on an olfactory short-term memory task. Front Behav Neurosci. 2012;6:59.PubMedPubMedCentral Devore S, Manella LC, Linster C. Blocking muscarinic receptors in the olfactory bulb impairs performance on an olfactory short-term memory task. Front Behav Neurosci. 2012;6:59.PubMedPubMedCentral
263.
Zurück zum Zitat Oh E, Park J, Youn J, Kim JS, Park S, Jang W. Olfactory dysfunction in early Parkinson’s disease is associated with short latency afferent inhibition reflecting central cholinergic dysfunction. Clin Neurophysiol. 2017;128:1061–8.PubMedCrossRef Oh E, Park J, Youn J, Kim JS, Park S, Jang W. Olfactory dysfunction in early Parkinson’s disease is associated with short latency afferent inhibition reflecting central cholinergic dysfunction. Clin Neurophysiol. 2017;128:1061–8.PubMedCrossRef
264.
Zurück zum Zitat Versace V, Langthaler PB, Sebastianelli L, Höller Y, Brigo D, Orioli A, et al. Impaired cholinergic transmission in patients with Parkinson’s disease and olfactory dysfunction. J Neurol Sci. 2017;377:55–61.PubMedCrossRef Versace V, Langthaler PB, Sebastianelli L, Höller Y, Brigo D, Orioli A, et al. Impaired cholinergic transmission in patients with Parkinson’s disease and olfactory dysfunction. J Neurol Sci. 2017;377:55–61.PubMedCrossRef
265.
Zurück zum Zitat Smith RS, Hu R, DeSouza A, Eberly CL, Krahe K, Chan W, et al. Differential muscarinic modulation in the olfactory bulb. J Neurosci. 2015;35:10773–85.PubMedPubMedCentralCrossRef Smith RS, Hu R, DeSouza A, Eberly CL, Krahe K, Chan W, et al. Differential muscarinic modulation in the olfactory bulb. J Neurosci. 2015;35:10773–85.PubMedPubMedCentralCrossRef
266.
Zurück zum Zitat McLean JH, Shipley MT. Serotonergic afferents to the rat olfactory bulb: I. Origins and laminar specificity of serotonergic inputs in the adult rat. J Neurosci. 1987;7:3016–28.PubMedCrossRef McLean JH, Shipley MT. Serotonergic afferents to the rat olfactory bulb: I. Origins and laminar specificity of serotonergic inputs in the adult rat. J Neurosci. 1987;7:3016–28.PubMedCrossRef
267.
Zurück zum Zitat Brill J, Shao Z, Puche AC, Wachowiak M, Shipley MT. Serotonin increases synaptic activity in olfactory bulb glomeruli. J Neurophysiol. 2016;115:1208–19.PubMedPubMedCentralCrossRef Brill J, Shao Z, Puche AC, Wachowiak M, Shipley MT. Serotonin increases synaptic activity in olfactory bulb glomeruli. J Neurophysiol. 2016;115:1208–19.PubMedPubMedCentralCrossRef
268.
Zurück zum Zitat Qamhawi Z, Towey D, Shah B, et al. Clinical correlates of raphe serotonergic dysfunction in early Parkinson’s disease. Brain. 2015;138:2964–73.PubMedCrossRef Qamhawi Z, Towey D, Shah B, et al. Clinical correlates of raphe serotonergic dysfunction in early Parkinson’s disease. Brain. 2015;138:2964–73.PubMedCrossRef
269.
Zurück zum Zitat Vermeiren Y, Janssens J, Van Dam D, De Deyn PP. Serotonergic dysfunction in amyotrophic lateral sclerosis and Parkinson’s disease: similar mechanisms, dissimilar outcomes. Front Neurosci. 2018;12:185.PubMedPubMedCentralCrossRef Vermeiren Y, Janssens J, Van Dam D, De Deyn PP. Serotonergic dysfunction in amyotrophic lateral sclerosis and Parkinson’s disease: similar mechanisms, dissimilar outcomes. Front Neurosci. 2018;12:185.PubMedPubMedCentralCrossRef
270.
Zurück zum Zitat Kovacs CG, Klöppel S, Fischer I, Domer S, Lindeck-Pozza E, Birner P, et al. Nucleus-specific alteration of raphe neurons in human neurodegenerative disorders. Neuroreport. 2003;14:73–6.PubMedCrossRef Kovacs CG, Klöppel S, Fischer I, Domer S, Lindeck-Pozza E, Birner P, et al. Nucleus-specific alteration of raphe neurons in human neurodegenerative disorders. Neuroreport. 2003;14:73–6.PubMedCrossRef
271.
Zurück zum Zitat McKeith IG, Dickson DW, Lwe 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, Lwe J, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65:1863–72.PubMedCrossRef
272.
Zurück zum Zitat Gilbert PE, Barr PJ, Murphy C. Differences in olfactory and visual memory in patients with pathologically confirmed Alzheimer’s disease and the Lewy body variant of Alzheimer’s disease. J Int Neurospychol Soc. 2004;10:835–42. Gilbert PE, Barr PJ, Murphy C. Differences in olfactory and visual memory in patients with pathologically confirmed Alzheimer’s disease and the Lewy body variant of Alzheimer’s disease. J Int Neurospychol Soc. 2004;10:835–42.
273.
Zurück zum Zitat Funabe S, Takao M, Saito Y, et al. Neuropathologic analysis of Lewy-related α-synucleinopathy in olfactory mucosa. Neuropathology. 2013;33:47–58.PubMedCrossRef Funabe S, Takao M, Saito Y, et al. Neuropathologic analysis of Lewy-related α-synucleinopathy in olfactory mucosa. Neuropathology. 2013;33:47–58.PubMedCrossRef
274.
Zurück zum Zitat Hepp DH, Vergoossen DL, Huisman E, et al. Distribution and load of amyloid-b pathology in Parkinson disease and dementia with Lewy bodies. J Neuropathol Exp Neurol. 2016;75:936–45.PubMedCrossRef Hepp DH, Vergoossen DL, Huisman E, et al. Distribution and load of amyloid-b pathology in Parkinson disease and dementia with Lewy bodies. J Neuropathol Exp Neurol. 2016;75:936–45.PubMedCrossRef
275.
Zurück zum Zitat McKhann GM, Knopman DS, Cheretkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–9.PubMedPubMedCentralCrossRef McKhann GM, Knopman DS, Cheretkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–9.PubMedPubMedCentralCrossRef
276.
Zurück zum Zitat Zou YM, Lu D, Liu LP, Zhang HH, Zhou YY. Olfactory dysfunction in Alzheimer’s disease. Neuropsychiatry Dis Treat. 2016;12:869–75.CrossRef Zou YM, Lu D, Liu LP, Zhang HH, Zhou YY. Olfactory dysfunction in Alzheimer’s disease. Neuropsychiatry Dis Treat. 2016;12:869–75.CrossRef
277.
Zurück zum Zitat Sun GH, Raji CA, Maceachern P, Burke JF. Olfactory identification testing as a predictor of the development of Alzheimer’s dementia: a systematic review. Laryngoscope. 2012;122:1455–62.PubMedCrossRef Sun GH, Raji CA, Maceachern P, Burke JF. Olfactory identification testing as a predictor of the development of Alzheimer’s dementia: a systematic review. Laryngoscope. 2012;122:1455–62.PubMedCrossRef
278.
Zurück zum Zitat Bahar-Fuchs A, Chételat G, Villemagner VL, Moss S, Pike K, Masters CL, et al. Olfactory deficits and amyloid-β burden in Alzheimer’s disease, mild cognitive impairment, and healthy aging: a PIB PET study. J Alzheimers Dis. 2010;22:1081–7.PubMedCrossRef Bahar-Fuchs A, Chételat G, Villemagner VL, Moss S, Pike K, Masters CL, et al. Olfactory deficits and amyloid-β burden in Alzheimer’s disease, mild cognitive impairment, and healthy aging: a PIB PET study. J Alzheimers Dis. 2010;22:1081–7.PubMedCrossRef
279.
Zurück zum Zitat Kim JY, Rasheed A, Yoo SJ, Kim SY, Cho B, Son G, et al. Distinct amyloid precursor protein processing machineries of the olfactory system. Biochem Biophys Res Commun. 2018;495:533–8.PubMedCrossRef Kim JY, Rasheed A, Yoo SJ, Kim SY, Cho B, Son G, et al. Distinct amyloid precursor protein processing machineries of the olfactory system. Biochem Biophys Res Commun. 2018;495:533–8.PubMedCrossRef
280.
Zurück zum Zitat Serby M, Larson P, Kalkstein D. The nature and course of olfactory deficits in Alzheimer’s disease. Am J Psychiatry. 1991;148:357–60.PubMedCrossRef Serby M, Larson P, Kalkstein D. The nature and course of olfactory deficits in Alzheimer’s disease. Am J Psychiatry. 1991;148:357–60.PubMedCrossRef
281.
Zurück zum Zitat Velayudhan L, Pritchard M, Powell JF, Proitsi P, Lovestone S. Smell identification function as a severity and progression marker in Alzheimer’s disease. Int Psychogeriatr. 2013;25:1157–66.PubMedCrossRef Velayudhan L, Pritchard M, Powell JF, Proitsi P, Lovestone S. Smell identification function as a severity and progression marker in Alzheimer’s disease. Int Psychogeriatr. 2013;25:1157–66.PubMedCrossRef
282.
Zurück zum Zitat Djordjevic J, Jones-Gotman M, De Sousa K, Chertkow H. Olfaction in patients with mild cognitive impairment and Alzheimer’s disease. Neurobiol Aging. 2008;29:693–706.PubMedCrossRef Djordjevic J, Jones-Gotman M, De Sousa K, Chertkow H. Olfaction in patients with mild cognitive impairment and Alzheimer’s disease. Neurobiol Aging. 2008;29:693–706.PubMedCrossRef
283.
Zurück zum Zitat Wilson RS, Arnold SE, Schneider JA, Boyle PA, Buchman AS, Bennett DA. Olfactory impairment in presymptomatic Alzheimer’s disease. Ann NY Acad Sci. 2009;1170:730–5.PubMedCrossRef Wilson RS, Arnold SE, Schneider JA, Boyle PA, Buchman AS, Bennett DA. Olfactory impairment in presymptomatic Alzheimer’s disease. Ann NY Acad Sci. 2009;1170:730–5.PubMedCrossRef
284.
Zurück zum Zitat Devanand DP, Michaels-Marston KS, Liu X, Pelton GH, Padilla M, Marder K, et al. Olfactory deficits in patients with mild cognitive impairment predict Alzheimer’s disease at follow-up. Am J Psychiatry. 2000;157:1399–405.PubMedCrossRef Devanand DP, Michaels-Marston KS, Liu X, Pelton GH, Padilla M, Marder K, et al. Olfactory deficits in patients with mild cognitive impairment predict Alzheimer’s disease at follow-up. Am J Psychiatry. 2000;157:1399–405.PubMedCrossRef
285.
Zurück zum Zitat Bahar-Fuchs A, Moss S, Rowe C, Savage G. Awareness of olfactory deficits in healthy aging, amnestic mild cognitive impairment and Alzheimer’s disease. Int Psychogeriatr. 2011;23:1097–106.PubMedCrossRef Bahar-Fuchs A, Moss S, Rowe C, Savage G. Awareness of olfactory deficits in healthy aging, amnestic mild cognitive impairment and Alzheimer’s disease. Int Psychogeriatr. 2011;23:1097–106.PubMedCrossRef
286.
Zurück zum Zitat Devanand DP, Lee S, Manly J, et al. Olfactory deficits predict cognitive decline and Alzheimer dementia in an urban community. Neurology. 2015;84:182–9.PubMedPubMedCentralCrossRef Devanand DP, Lee S, Manly J, et al. Olfactory deficits predict cognitive decline and Alzheimer dementia in an urban community. Neurology. 2015;84:182–9.PubMedPubMedCentralCrossRef
287.
Zurück zum Zitat Doty RL, Reyes PF, Gregor T. Presence of both odor identification and detection deficits in Alzheimer’s disease. Brain Res Bull. 1987;18:597–600.PubMedCrossRef Doty RL, Reyes PF, Gregor T. Presence of both odor identification and detection deficits in Alzheimer’s disease. Brain Res Bull. 1987;18:597–600.PubMedCrossRef
288.
Zurück zum Zitat Conti MZ, Vicini-Chilovi B, Riva M, Zanetti M, Liberini P, Padovani A, et al. Odor identification deficit predicts clinical conversion from mild cognitive impairment to dementia due to Alzheimer’s disease. Arch Clin Neuropsychol. 2013;28:391–9.PubMedCrossRef Conti MZ, Vicini-Chilovi B, Riva M, Zanetti M, Liberini P, Padovani A, et al. Odor identification deficit predicts clinical conversion from mild cognitive impairment to dementia due to Alzheimer’s disease. Arch Clin Neuropsychol. 2013;28:391–9.PubMedCrossRef
289.
Zurück zum Zitat Devanand DP, Liu X, Tabert MH, et al. Combining early markers strongly conversion from mild cognitive impairment to Alzheimer’s disease. Biol Psychiatr. 2008;64:871–9.CrossRef Devanand DP, Liu X, Tabert MH, et al. Combining early markers strongly conversion from mild cognitive impairment to Alzheimer’s disease. Biol Psychiatr. 2008;64:871–9.CrossRef
290.
Zurück zum Zitat Murphy C, Jernigan TL, Fennema-Notestine C. Left hippocampal volume loss in Alzheimer’s disease is reflected in performance on odor identification: a structural MRI study. J Int Neuropsychol Soc. 2003;9:459–71.PubMedCrossRef Murphy C, Jernigan TL, Fennema-Notestine C. Left hippocampal volume loss in Alzheimer’s disease is reflected in performance on odor identification: a structural MRI study. J Int Neuropsychol Soc. 2003;9:459–71.PubMedCrossRef
291.
292.
Zurück zum Zitat Kovacs T, Cairns NJ, Lantos PL. Beta-amyloid deposition and neurofibrillary tangle formation in the olfactory bulb in ageing and Alzheimer’s disease. Neuropathol Appl Neurobiol. 1999;25:481–91.PubMedCrossRef Kovacs T, Cairns NJ, Lantos PL. Beta-amyloid deposition and neurofibrillary tangle formation in the olfactory bulb in ageing and Alzheimer’s disease. Neuropathol Appl Neurobiol. 1999;25:481–91.PubMedCrossRef
293.
Zurück zum Zitat Wesson DW, Levy E, Nixon RA, Wilson DA. Olfactory dysfunction correlates with amyloid-beta burden in an Alzheimer’s disease mouse model. J Neurosci. 2010;30:505–14.PubMedPubMedCentralCrossRef Wesson DW, Levy E, Nixon RA, Wilson DA. Olfactory dysfunction correlates with amyloid-beta burden in an Alzheimer’s disease mouse model. J Neurosci. 2010;30:505–14.PubMedPubMedCentralCrossRef
294.
Zurück zum Zitat Xu W, Fitzgerald S, Nixon RA, Levy E, Wilson DA. Early hyperactivity in lateral entorhinal cortex is associated with elevated levels of AβPP metabolites in the Tg2576 mouse model of Alzheimer’s disease. Exp Neurol. 2015;264:82–91.PubMedCrossRef Xu W, Fitzgerald S, Nixon RA, Levy E, Wilson DA. Early hyperactivity in lateral entorhinal cortex is associated with elevated levels of AβPP metabolites in the Tg2576 mouse model of Alzheimer’s disease. Exp Neurol. 2015;264:82–91.PubMedCrossRef
295.
Zurück zum Zitat Hu B, Geng C, Hou XY. Oligomeric amyloid-b peptide disrupts olfactory information output by impairment of local inhibitory circuits in rat olfactory bulb. Neurobiol Aging. 2017;51:113–21.PubMedCrossRef Hu B, Geng C, Hou XY. Oligomeric amyloid-b peptide disrupts olfactory information output by impairment of local inhibitory circuits in rat olfactory bulb. Neurobiol Aging. 2017;51:113–21.PubMedCrossRef
296.
Zurück zum Zitat •• Risacher SL, Tallman EF, West JD, et al. Olfactory identification in subjective cognitive decline and mild cognitive impairment: association with tau but not amyloid positron emission tomography. Alzheimers Dement. 2017;9:57–66. Using amyloid positron emission tomography, magnetic resonance and UPSIT test, the present report demonstrates that olfactory deficit may be a good marker for tau and neurodegeneration in preclinical or prodromal Alzheimer’s disease. •• Risacher SL, Tallman EF, West JD, et al. Olfactory identification in subjective cognitive decline and mild cognitive impairment: association with tau but not amyloid positron emission tomography. Alzheimers Dement. 2017;9:57–66. Using amyloid positron emission tomography, magnetic resonance and UPSIT test, the present report demonstrates that olfactory deficit may be a good marker for tau and neurodegeneration in preclinical or prodromal Alzheimer’s disease.
297.
Zurück zum Zitat Attems J, Jellinger KA. Olfactory tau pathology in Alzheimer disease and mild cognitive impairment. Clin Neuropathol. 2006;25:265–71.PubMed Attems J, Jellinger KA. Olfactory tau pathology in Alzheimer disease and mild cognitive impairment. Clin Neuropathol. 2006;25:265–71.PubMed
298.
Zurück zum Zitat Goedert M. The ordered assembly of tau is the gain-of-toxic function that causes human tauopathies. Alzheimers Dement. 2016;12:1040–50.PubMedCrossRef Goedert M. The ordered assembly of tau is the gain-of-toxic function that causes human tauopathies. Alzheimers Dement. 2016;12:1040–50.PubMedCrossRef
299.
Zurück zum Zitat Oleson S, Murphy C. Olfactory dysfunction in ApoE E4/4 homozygotes with Alzheimer’s disease. J Alzheimers Dis. 2015;46:791–803.PubMedCrossRef Oleson S, Murphy C. Olfactory dysfunction in ApoE E4/4 homozygotes with Alzheimer’s disease. J Alzheimers Dis. 2015;46:791–803.PubMedCrossRef
300.
Zurück zum Zitat Juottonen K, Lehtovirta M, Helisalmi S, Riekkinen PJ Sr, Soininen H. Major decrease in the volume of the entorhinal cortex in patients with Alzheimer’s disease carrying the apolipoprotein E epsilon 4 allele. J Neurol Neurosurg Psychiatry. 1998;65:322–7.PubMedPubMedCentralCrossRef Juottonen K, Lehtovirta M, Helisalmi S, Riekkinen PJ Sr, Soininen H. Major decrease in the volume of the entorhinal cortex in patients with Alzheimer’s disease carrying the apolipoprotein E epsilon 4 allele. J Neurol Neurosurg Psychiatry. 1998;65:322–7.PubMedPubMedCentralCrossRef
301.
Zurück zum Zitat Olofsson JK, Rönnlund M, Nordin S, Nyberg L, Nilsson LG, Larsson M. Odor identification deficit as a predictor of five-year global cognitive change: interactive effects with age and ApoE-epsilon4. Behav Genet. 2009;39:496–503.PubMedCrossRef Olofsson JK, Rönnlund M, Nordin S, Nyberg L, Nilsson LG, Larsson M. Odor identification deficit as a predictor of five-year global cognitive change: interactive effects with age and ApoE-epsilon4. Behav Genet. 2009;39:496–503.PubMedCrossRef
302.
Zurück zum Zitat Devanand DP, Tabert MH, Cuasay K, et al. Olfactory identification deficits and MCI in a multi-ethnic elderly community sample. Neurobiol Aging. 2010;31:1593–600.PubMedCrossRef Devanand DP, Tabert MH, Cuasay K, et al. Olfactory identification deficits and MCI in a multi-ethnic elderly community sample. Neurobiol Aging. 2010;31:1593–600.PubMedCrossRef
303.
Zurück zum Zitat Lehéricy S, Hirsch EC, Cervera-Piérot P, et al. Heterogeneity and selectivity of the degeneration of cholinergic neurons in the basal forebrain of patients with Alzheimer’s disease. J Comp Neurol. 1993;330:15–31.PubMedCrossRef Lehéricy S, Hirsch EC, Cervera-Piérot P, et al. Heterogeneity and selectivity of the degeneration of cholinergic neurons in the basal forebrain of patients with Alzheimer’s disease. J Comp Neurol. 1993;330:15–31.PubMedCrossRef
304.
Zurück zum Zitat Tiraboschi P, Hansen LA, Alford M, Merdes A, Masliah E, Thal LJ, et al. Early and widespread cholinergic losses differentiate dementia with Lewy bodies from Alzheimer disease. Arch Gen Psychiatry. 2002;59:946951.CrossRef Tiraboschi P, Hansen LA, Alford M, Merdes A, Masliah E, Thal LJ, et al. Early and widespread cholinergic losses differentiate dementia with Lewy bodies from Alzheimer disease. Arch Gen Psychiatry. 2002;59:946951.CrossRef
305.
Zurück zum Zitat Ahlskog JE, Waring SC, Petersen RC, et al. Olfactory dysfunction in Guamanian ALS, parkinsonism, and dementia. Neurology. 1998;51:1672–7.PubMedCrossRef Ahlskog JE, Waring SC, Petersen RC, et al. Olfactory dysfunction in Guamanian ALS, parkinsonism, and dementia. Neurology. 1998;51:1672–7.PubMedCrossRef
306.
Zurück zum Zitat Günther R, Schrempf W, Häher A, Hummel T, Wolz M, Storch A, et al. Impairment in respiratory function contributes to olfactory impairment in amyotrophic lateral sclerosis. Front Neurosci. 2018;9:79.CrossRef Günther R, Schrempf W, Häher A, Hummel T, Wolz M, Storch A, et al. Impairment in respiratory function contributes to olfactory impairment in amyotrophic lateral sclerosis. Front Neurosci. 2018;9:79.CrossRef
307.
Zurück zum Zitat Hawkes CH, Shepard BC, Geddes JF, Body GD, Martin JE. Olfactory disorder in motor neuron disease. Exp Neurol. 1998;150:248–53.PubMedCrossRef Hawkes CH, Shepard BC, Geddes JF, Body GD, Martin JE. Olfactory disorder in motor neuron disease. Exp Neurol. 1998;150:248–53.PubMedCrossRef
308.
Zurück zum Zitat Pilotto A, Rossi F, Rinaldi F, et al. Exploring olfactory function and its relation with behavioural and cognitive impairment in amyotrophic lateral sclerosis patients: a cross-sectional study. Neurodegener Dis. 2016;16:411–6.PubMedCrossRef Pilotto A, Rossi F, Rinaldi F, et al. Exploring olfactory function and its relation with behavioural and cognitive impairment in amyotrophic lateral sclerosis patients: a cross-sectional study. Neurodegener Dis. 2016;16:411–6.PubMedCrossRef
309.
Zurück zum Zitat Eisen A, Braak H, Tredici KD, Lemon R, Ludolph AC, Kiernan MC. Cortical influences drive amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry. 2017;88:917–24.PubMedCrossRef Eisen A, Braak H, Tredici KD, Lemon R, Ludolph AC, Kiernan MC. Cortical influences drive amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry. 2017;88:917–24.PubMedCrossRef
Metadaten
Titel
Olfactory Dysfunction in Neurodegenerative Diseases
verfasst von
Concepció Marin
Dolores Vilas
Cristóbal Langdon
Isam Alobid
Mauricio López-Chacón
Antje Haehner
Thomas Hummel
Joaquim Mullol
Publikationsdatum
01.08.2018
Verlag
Springer US
Erschienen in
Current Allergy and Asthma Reports / Ausgabe 8/2018
Print ISSN: 1529-7322
Elektronische ISSN: 1534-6315
DOI
https://doi.org/10.1007/s11882-018-0796-4

Weitere Artikel der Ausgabe 8/2018

Current Allergy and Asthma Reports 8/2018 Zur Ausgabe

Anaphylaxis and Drug Allergy (DA Khan and M Castells, Section Editors)

Food-Induced Anaphylaxis: an Update

Pediatric Allergy and Immunology (W Dolen, Section Editor)

The Child with Recurrent Mycobacterial Disease

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Hörschwäche erhöht Demenzrisiko unabhängig von Beta-Amyloid

29.05.2024 Hörstörungen Nachrichten

Hört jemand im Alter schlecht, nimmt das Hirn- und Hippocampusvolumen besonders schnell ab, was auch mit einem beschleunigten kognitiven Abbau einhergeht. Und diese Prozesse scheinen sich unabhängig von der Amyloidablagerung zu ereignen.

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Betalaktam-Allergie: praxisnahes Vorgehen beim Delabeling

16.05.2024 Pädiatrische Allergologie Nachrichten

Die große Mehrheit der vermeintlichen Penicillinallergien sind keine. Da das „Etikett“ Betalaktam-Allergie oft schon in der Kindheit erworben wird, kann ein frühzeitiges Delabeling lebenslange Vorteile bringen. Ein Team von Pädiaterinnen und Pädiatern aus Kanada stellt vor, wie sie dabei vorgehen.

Update HNO

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.