Abstract
This review summarizes major advances in biomarker discovery for diagnosis, differential diagnosis, and progression of Parkinson’s disease (PD), with emphasis on neuroimaging and biochemical markers. Potential strategies to develop biomarkers capable of predicting PD in the prodromal stage before the appearance of motor symptoms or correlating with nonmotor symptoms, an active area of research, are also discussed.
Keywords: Parkinson disease, biomarkers, neuroimaging, proteomics, metabolomics, prodromal diagnosis
Parkinson’s disease (PD) is a prevalent neurodegenerative disorder, with its motor symptoms largely attributable to the degeneration of dopaminergic (DAergic) neurons. It has become clear, however, that PD is also associated with nonmotor symptoms in conjunction with neuronal loss in many other brain regions that occur before or after the loss of DAergic neurons [1]. The misdiagnosis rate of PD can range from 10–50% by movement disorder specialists [2] due in part to the fact that there are no sensitive and specific biomarkers validated for clinicians to differentiate PD from other movement disorders with overlapping clinical symptoms. Apart from diagnostic utility, biomarkers for PD are also needed for monitoring disease progression and efficacy of interventions, which are currently assessed by severity of motor symptoms.
An ideal PD biomarker should meet the following qualifications: high sensitivity and specificity validated by neuropathological examination, satisfactory test–retest reproducibility, easy accessibility, inexpensive, and offer the ability to monitor disease progression without being biased by age, compensatory mechanisms, or treatments. While no such biomarker to date fulfills all of these criteria for PD, this review will discuss two areas of research, neuroimaging and biochemical markers, which have demonstrated obvious potential in diagnosing the disease and monitoring its progression. Future biomarker investigations relating to prodromal biomarkers and markers related to nonmotor symptoms are also discussed.
Neuroimaging biomarkers
To date, the most mature PD biomarkers for nigrostriatal neurodegeneration are those employing neuroimaging methodologies. Currently, controversy surrounds whether these techniques can be effective in differentiating clinically overlapping parkinsonisms and/or objectively assessing PD progression. The major methods used as well as their current clinical and research utility are discussed below and summarized in Table 1.
Table 1.
Neuroimaging biomarker candidates and their potential utilities in PD.
| Imaging target | Prodromal diagnosis | Confirming PD at its onset | Differentiation from atypical Parkinsonism | Monitoring progression | Sensitivity and specificity for PD diagnosis [Refs] |
|---|---|---|---|---|---|
| AADC (18F-dopa) | Probably useful | Useful | Unlikely useful | Possibly useful | |
| DAT | Useful | Useful | Unlikely useful | Possibly useful | 98% and 83%[86], 92% and 100% [18] |
| VMAT2 | Probably useful | Useful | Investigational | Investigational | |
| FDG network | Probably useful | Probably useful | Probably useful | Possibly useful | 95% and 94% [87] |
| Dopamine receptors | Non-useful | Non-useful alone | Non-useful alone | Non-useful | |
| Cholinergic function | Investigational | Investigational | Probably useful (for DLB) | Investigational | |
| Cardiac autonomic dysfunction | Investigational | Non-useful | Useful (for MSA) | Investigational | |
| TCS | Probably useful | Useful | Investigational | Unlikely useful | 91% and 82% [68] |
| MRI (DWI, iron deposit) | Investigational | Probably useful | Investigational | Investigational |
Dopaminergic imaging
Aromatic amino acid decarboxylase
6-[18F]-fluoro-l-3,4-dihydroxyphenylalanine (18F-dopa) positron emission tomography (PET) was the first neuroimaging approach validated for measuring and assessing presynaptic DAergic neuronal integrity. To reach the striatum, 18F-dopa is transported across the blood–brain barrier, taken up by axonal terminals of DA neurons, decarboxylated by aromatic amino acid decarboxylase (AADC), converted into 18F-DA, and stored in vesicles. The key determinants for 18F-dopa uptake are the density of the axonal terminal plexus and AADC activity, which reflect the number of remaining nigral DAergic cells. Patients with early PD show 50% decreased 18F-dopa uptake in the putamen [3], with the greatest reduction occurring in a gradient pattern in the posterior dorsal putamen contralateral to the side of symptom onset [4,5]. Although such imaging appears to be effective in confirming symptomatic PD, its utility in assessing PD progression [6,7] and detecting prodromal PD are questionable because 18F-dopa PET may underestimate the degree of degeneration due to compensatory upregulation of AADC in remaining terminals [8].
Dopamine transporter
The dopamine transporter (DAT), a protein expressed on the membrane of presynaptic DA terminals and involved in the reuptake of DA, can be assessed with PET using several tracers, including 11C-CFT, 18F-FP-CIT, 11C-RTI-32, and 11C-methylphenidate. Single photon emission computed tomography (SPECT), a technology used routinely in clinical practice, can also be used to image DAT through 123I-b-CIT, 123I-FP-CIT, 123I-IPT, 123I-altropane, or 99mTc- TRODAT-1 [9]. DAT imaging is used as an in vivo marker, reflecting integrity and number of DA neurons. A few studies have reported significantly reduced striatal DAT in more than 95% of parkinsonism cases [10], including those at early stages [11].
With respect to monitoring PD progression and efficacy of putative agents, the value of DAT imaging remains to be established. Although some investigations suggest that striatal uptake of 18F-FP-CIT correlates with Hoehn and Yahr (H&Y) score [12], other clinical trials show that DAT imaging density declined faster in patients with PD treated with levodopa, despite improved clinical motor scores [13]. Symptomatic therapy may therefore influence imaging results in their relation to clinical diagnosis.
Vesicular monoamine transporter 2
Vesicular monoamine transporter 2 (VMAT2), a membrane protein that transports monoamines from the cytosol into secretory vesicles in monoaminergic neurons, is exclusively expressed in the brain and has an essential role in DA reuptake. VMAT2 is imaged using 11C- or 18F-dihydrotetrabenazine (DTBZ) PET and is the newest approach for the assessment of nigrostriatal projections [14]. In patients with PD, striatal 11C-DTBZ is significantly reduced with the putamen exhibiting the greatest decrease [14]. VMAT2 imaging appears to be less sensitive to compensation and pharmacologic regulation [15], giving it the potential to provide the most reliable measurement of the density of DAergic terminals in PD. Nevertheless, the application of 11C-DTBZ is limited because its short half-life requires a cyclotron on-site [16]. A novel 18F-labeled tetrabenazine derivative, 18F-fluoropropyldihydrotetrabenazine, is currently under development and is showing promise as a longer-lived and lower-cost alternative [16].
Caveats and potential solutions
Among the three techniques discussed above, DAT imaging is the most commonly used to date. However, several limitations associated with its use should be emphasized. DAT imaging likely overestimates the reduction in terminal density in early PD due to compensatory downregulation in remaining neurons. Also, current DAT radioligands bind with other monoamine transporters, particularly serotonin (SERT), which is especially problematic as SERT is the dominant monoamine transporter in the midbrain [17]. Lastly, DAT imaging cannot reliably differentiate between PD and other forms of parkinsonism [18], making the production of additional methods in conjunction with DAT imaging necessary to define a PD biomarker. Recently, fluorine-18 labeled 2-βcarbomethoxy-3β-(4-chlorophenyl)-8-(2-fluoroethyl)-nortropane (18F-FECNT) was developed as a novel PET tracer with high affinity for DAT and a much lower affinity for the norepinephrine transporter and SERT. Compared with current PET DAT tracers, 18F-FECNT has higher test–retest reproducibility and may be able to track striatal and nigral DA denervation [19], making it potentially useful for longitudinal evaluation of PD progression.
As previously mentioned, another shortcoming of DAergic neuroimaging is that the techniques assess nigrostriatal DA function, rather than true pathology, rendering the obtained values subject to compensatory mechanisms, in which there is upregulation of 18F-dopa uptake and downregulation of DAT binding [15]. To partially circumvent this problem, postsynaptic DA receptors can be examined by the PET ligand 11C-raclopride for D2/3 receptor [20] or SPECT tracer 123I-iodobenzamide for D2 receptors. Increased levels of D2 receptor availability can be observed in early stages of de novo PD, which is useful in differentiating PD from atypical parkinsonisms [21]. Nonetheless, whether these new targets are truly unaffected by compensatory modulations or medications remains to be investigated.
Non-dopaminergic imaging
An active area of research in neuroimaging markers is assessment of brain functions and structures beyond nigrostriatal DAergic degeneration. These include Parkinson disease-related spatial covariance pattern (PDRP), cholinergic function imaging, and magnetic resonance imaging (MRI). Such strategies incorporate the fact that PD affects non-nigrostriatal DAergic regions of the brain. Transcranial sonography (TCS), also a non-dopamine imaging method, will be discussed in the context of prodromal diagnosis, where utility of imaging methods will be integrated with biochemical markers.
Metabolism network imaging in brain
Investigation with 18F-FDG PET revealed that PD is associated with a specific metabolic network characterized by increased pallidothalamic and pontine metabolism associated with metabolic reductions in the lateral premotor and posterior parietal cortical regions [22,23], termed PDRP. In one study, PDRP network analysis classified idiopathic PD with 84% sensitivity and 97% specificity, helping differentiate PD from atypical parkinsonisms such as multiple system atrophy (MSA) and progressive supranuclear palsy (PSP) [24]. Furthermore, PDRP activity precedes the appearance of motor symptoms by approximately 2 years [25], indicating its potential usefulness in diagnosing PD at its prodromal stage. These facts, coupled with its high level of reproducibility [26] and correlation with clinical severity [27], make PDRP a promising biomarker to track PD progression, monitor therapeutic intervention [28], and could potentially fill voids left by nigrostriatal imaging methods mentioned above.
Cholinergic dysfunction imaging
In addition to PDRP, imaging brain regions that are not part of the nigrostriatal tract shows some promise in diagnosing and distinguishing PD from other neurological diseases. New imaging techniques for assaying cortical acetylcholinesterase activity with 11C-MP4A PET reveal a deficit of cholinergic function throughout the cortex in parallel with the loss of striatal DAergic function in PD [29]. This observation is significant, especially with the realization of nonmotor components in PD (e.g. cognitive impairment) which is intimately associated with the cholinergic system.
MRI
MRI is a form of neuroimaging that is particularly useful in ruling out secondary causes of parkinsonism due to the fact that it is capable of detecting abnormalities in the structure of various brain regions. Recent advances in high-field MRI technology have been increasingly employed in diagnosing PD and are more sensitive towards demonstrating iron deposits in the midbrain of early PD patients [30]. Additionally, diffusion tensor imaging (DTI) for evaluating regional fractional anisotropy has shown changes in the olfactory tract [31], which could be related to anosmia or hyposmia (a symptom that appears in the prodromal stage – see discussion below), and reduction in the nigra of patients with PD [32] which inversely correlates with H&Y score [33].
Biochemical biomarkers
Biochemical markers, especially those reflecting PD pathogenesis, are sorely needed in samples that are readily accessible clinically [e.g. cerebrospinal fluid (CSF), blood, and saliva]. To date, the most extensively tested candidate biochemical markers are those present in CSF and typically stem from genetic investigations which link the candidate to PD development. A few major biochemical markers and their current clinical and research utility are listed in Table 2. However, none of these markers completely fulfill the criteria defined earlier as an ideal biomarker for PD. Most new candidate markers are discovered by ‘-omics’ technology, and are generally in a preliminary stage with the results obtained in small cohorts using cross-sectional study designs. Validation is therefore needed using large cohorts, particularly those with samples collected longitudinally. Finally, similar to the neuroimaging field, major challenges to the biochemical marker field also include defining prodromal biomarkers and those related to nonmotor dysfunctions.
Table 2.
Biochemical Biomarker candidates and their potential utilities in PD.
| Biochemical marker(s) | Prodromal diagnosis [Refs] | Confirming PD at its onset [Refs] | Differentiation from atypical Parkinsonism [Refs] | Monitoring progression [Refs] | Sensitivity and specificity for PD diagnosis [Refs] |
|---|---|---|---|---|---|
| α-Synuclein | Unlikely in LRRK2 cases [83] | Possibly useful [36–39] | Possibly useful-potentially more useful if PS-129 is measured or it is used with the ratio of p-tau/tau [42], | Possibly useful [42] | Measuring oligomers 75% and 87.5%; improves to 89.3% and 90.6% when measuring the ratio of oligomers:total [40] |
| DJ-1 | Possibly useful in G2019S LRRK2 cases [83] | Possibly useful [43] | Unknown | Possibly useful [43] | |
| Aβ | Possibly useful [83] | Possibly useful [48–50] | Unknown | Possibly useful if used in conjunction with other protein(s), e.g. tau, fractalkine [48–50]. | |
| Tau | Possibly useful [83] | Possibly useful [48,49] | Possibly useful if used in a p-tau/tau ratio with α-synuclein [42] | Possibly useful if used in conjunction with Aβ [48,50,51] | |
| Uric Acid | Unknown | Possibly useful [52] | Unknown | Possibly useful [55] | |
| Glutathione | Unknown | Possibly useful [52, 53] | Unknown | Unknown |
α-Synuclein, DJ-1, and tau
α-Synuclein has been intensely researched as a PD biomarker due to the fact that it is a key protein in Lewy bodies, the pathological hallmark of PD, and mutations or multiplication of its gene are known to cause familial PD [34,35]. α-Synuclein has been reported to be decreased in CSF from patients with PD compared with controls in most well-controlled investigations [36–39], although it does not appear to be able to distinguish between various synucleinopathies independently [39]. One study has reported an inverse relationship between α-synuclein levels and H&Y score [36], indicating measuring α-synuclein levels may not only be useful as a biomarker of PD but also of disease progression. However, the latter observation has not been replicated in most recent studies [38]. Additionally, oligomeric forms of α-synuclein have been found to be elevated in the CSF of patients with PD as compared with controls. When only oligomers were measured, the sensitivity and specificity were calculated to be 75% and 87.5%, respectively, which increases to 89.3% and 90.6% when the ratio of oligomers and/or total α-synuclein is calculated [40]. Similar elevations have also been observed in plasma [41]. A more recent study reports that the CSF level of phosphorylated α-synuclein (PS-129) appears to be more effective than native α-synuclein in differentiating PD from MSA and PSP and correlates with disease severity as assessed using the Unified Parkinson’s Disease Rating Scale (UPDRS) [42], making it a potential candidate to complement the neuroimaging methods discussed above.
DJ-1 is an extensively studied antioxidative protein which, like α-synuclein, is implicated in PD pathogenesis as mutation of its PARK7 gene results in familial autosomal recessive forms of the disease. Some reports indicate that DJ-1 is increased in CSF from patients with PD compared to controls [43], a finding which was replicated in plasma and extended to show that DJ-1 levels correlate with disease stage as approximated by H&Y scores [44]. Other studies, however, contradict this conclusion by showing DJ-1 to be decreased in CSF from patients with PD compared with controls, and show an apparent age-dependent increase in DJ-1 levels [38]. The discrepancy might be related to methodological variations or contamination of CSF by blood, which contains a comparatively high level of DJ-1 [45].
In addition to α-synuclein [46,47], genome-wide association studies have found associations between the gene encoding tau (MAPT) and PD. However, this observation has largely arisen in cohorts of European descent [46] and was not found in a Japanese cohort, indicating there may be geographical differences in genes associated with PD [47]. Studies with larger cohorts have reported that CSF levels of tau and phospho-181 tau (p-tau) are decreased in symptomatic PD compared with controls [48,49].
In addition to the cardinal motor indicators of PD, patients also experience significant nonmotor symptoms. As such, biomarker investigations have been launched in an attempt to characterize these effects. One such symptom is cognitive impairment, for which a decrease in CSF amyloid-β (Aβ) levels has been well established in patients with Alzheimer disease (AD). In CSF samples obtained from patients with PD, the Aβ isoforms Aβ40 and Aβ42 have been found to be decreased as compared with controls. However, in contrast to AD, there is no consistent increase in tau in conjunction with decreased Aβ in patients with PD with cognitive impairment or dementia [48,50,51].
It should be pointed out that due to the heterogeneity of patients with PD, a combination of several markers may be necessary to achieve high sensitivity and specificity. This is evidenced by the fact that the ratio of α-synuclein to the percentage of p-tau (p-tau/tau) is able to distinguish PD from MSA [49]. Furthermore, the ratio of fractalkine, an inflammatory mediator of microglia, to Aβ42 positively correlates with PD severity and progression in cross-sectional and longitudinal CSF samples, respectively [49]. Using combinations of protein markers and/or imaging techniques could therefore produce a biomarker that not only diagnoses PD, but also monitors disease progression. Alternatively, markers may also be stratified based on the predominant features of PD, such as tremor or rigidity, or at-risk traits and/or clinical features (discussed below).
Use of ‘omics’ in biomarker development
A variety of profiling techniques have been employed to find novel markers that could facilitate diagnosis and monitoring of PD progression. Indeed, the use of ‘-omics’ technologies have enabled for high-throughput studies of metabolites, genes, and proteins in the context of comparing patients with PD with controls. This section will discuss the major findings of metabolomic, genomic, gene expression profiling, and proteomic studies aimed at developing PD biomarkers.
Metabolomics
Metabolomics is used to study the profile of small molecules and has been used in a limited capacity to observe differences in metabolites that may aid in PD diagnosis. Previous studies report decreased uric acid and increased glutathione in plasma from patients with PD compared with controls [52]. However, the reduced form of glutathione has been reported to be decreased in CSF of patients with Lewy body disease, with patients with PD showing a non-statistically significant decrease of this metabolite [53]. By contrast, decreased plasma levels of uric acid have been found in both idiopathic PD and PD caused by mutations in leucine rich repeat kinase 2 (LRRK2), indicating decreased uric acid levels may be a universal feature of patients with PD [54]. Given both higher serum and CSF urate concentrations at baseline are associated with slower rates of clinical deterioration [55] and serum urate is reported to decrease with disease progression as measured by H&Y score [56], urate may be important in predicting and monitoring PD. Notably, the LRRK2 study discussed above also calculated the scores of projection to latent structures-discriminant analysis (PLS-DA) for related and unrelated controls in addition to for patients with idiopathic and LRRK2 PD and found that each form was distinguishable from the control in addition to each other, although common signatures are also noted. Another study using PLS-DA analysis has identified pyruvate as a key metabolite that can distinguish patients with PD from controls [57]. While metabolomic studies may provide a biomarker that can accurately diagnose PD, the limited number of published studies highlights the need for validation as well as comparisons of PD to other neurodegenerative diseases.
Genomic and expression profiles
A majority of the genomic work done relating to PD biomarker research has found the genes SNCA and MAPT, encoding for α-synuclein and tau, respectively, are associated with PD as described above [46,47]. A variety of studies have also examined differences in mRNA expression between PD and control subjects. Unfortunately, most of these have been performed in brain tissue, making them impractical towards the development of a clinically useful biomarker. There have been a select number of studies that measured transcript differences in blood. One such study looked at the blood mRNA signature and reported a molecular marker of eight genes that are indicative of a greater risk of PD, in particular decreased expression of ST13 [58]. Results for the expression of ST13, however, were not replicated in a separate study [59] and no differences were observed in a similar study of GSK3B in blood [60]. By contrast, several other genes discovered initially, including HIP2 and HSPA9 [58], appear to be reproducible in other investigations [61,62].
Proteomic profiling
Many studies have profiled various proteomes of PD and other neurodegenerative patients with the goal of detecting differences in proteins between these groups. A commonly used biofluid is CSF, because it is in close contact with the location of primary PD pathology and can be reasonably obtained. One such study used proteomic profiling of CSF from patients with PD, AD, and dementia with Lewy bodies and found unique changes for each group compared with healthy controls among the 1500 (approximately) proteins identified [63]. Additionally, each group was distinguishable from one another with 95% sensitivity achieved [63]. Later studies validated that levels of brain-derived neurotrophic factor, interleukin 8, vitamin D binding protein, β2-microglobulin, haptoglobin, apolipoprotein AII, apoE, tau, and Aβ42 could accurately classify 90 of 95 healthy controls, 36 of 48 AD patients, and 38 of 40 PD patients when compared with expert diagnosis [64]. This further supports the concept that using multiple proteins may be a key factor in developing a biomarker for PD. Other profiling studies have been performed and have found different candidate markers [65,66], illustrating that reproducibility is low when employing general profiling methods across diverse cohorts.
Future directions of biomarkers in PD
A biomarker that is able to diagnose PD before the onset of motor symptoms would provide a better chance to develop interventions capable of arresting or slowing disease progression. To this end, two types of high risk populations, subjects with genetic mutations leading to familial PD and those with clinical symptoms associated with a high conversion rate to PD, are candidates wherein prodromal biomarkers can be potentially identified. Additionally, discovery of biochemical markers is shifting from CSF to other peripheral biofluids that are more easily accessible. Imaging, when used in combination with biochemical biomarkers, may allow for the discovery of a specific profile that can predict PD onset.
Prodromal diagnosis
Tools used for prodromal diagnosis
The use of neuroimaging has shown the greatest promise in developing prodromal markers for PD. Nigrostriatal DA imaging, the best established marker of motor dysfunction thus far, should be considered the ‘gold-standard’ in defining subjects at a higher risk for PD before the onset of motor symptoms (discussed below). Additionally, TCS, which is readily available in most clinics, may be useful towards prodromal diagnosis of PD. To this end, TCS has been used to observe the lateral midbrain and studies report increased echogenicity (‘hyperechogenicity’) in the lateral midbrain in approximately 90% of cases [67]. Compared with the clinical standard, diagnosing PD at baseline by TCS was assessed with a sensitivity of 91% and specificity of 82% [68]. TCS may be useful in developing a prodromal biomarker as one study reports 14 of 39 patients with rapid eye movement behavior disorder (RBD), a group of people at risk of PD (discussed below), showed hyperechogenecity in the nigra [69]. Additionally, for individuals 50 years or older without evidence of PD, the relative risk for incident PD in those with enlarged substantia nigra hyperechogenicity was 17 times higher compared with normoechogenic controls after 37 months of prospective follow up [70]. It should be noted, however, the size of the TCS signal did not show changes with disease progression when assessed using H&Y or UPDRS scores [71], suggesting it is not an appropriate biomarker for follow-up evaluation of disease severity.
Population at high risk
To develop a prodromal biomarker for PD, individuals who are at risk of developing PD can be studied before disease onset. Two such populations include individuals who have a genetic risk for PD and people who show symptoms that appear before disease onset. Among individuals with a genetic risk for PD, those with mutations in LRRK2 or glucocerebrosidase (GBA) are worth discussing, because their prevalence is relatively high. Studies on asymptomatic LRRK2 carriers are likely to be especially informative due to the high penetrance rate of LRRK2 mutations [72]. DAT imaging of two asymptomatic LRRK2 mutation carriers showed reduced 11C-MP binding but normal 18F-dopa uptake in putamen [73], which is consistent with decreased binding of DAT and increased activity of dopa decarboxylase in prodromal stages of PD. Additionally, LRRK2 patients were similar to patients with sporadic PD [73]. A separate study reported a greater than expected decline in PET markers (most commonly 11C-DTBZ and 11C-methylphenidate but also 18F-dopa uptake) for some non-symptomatic LRRK2 carriers [74], indicating PET imaging before clinical PD onset may be useful in diagnosing PD. Others have shown that GBA mutation carriers have decreased cerebral rates of glucose metabolism in the supplemental motor area [75]; further validating the concept that imaging before the appearance of motor symptoms may be an important aspect of a prodromal PD biomarker.
Imaging subjects displaying nonmotor symptoms that commonly occur before the onset of motor symptoms in PD may also be useful in defining a prodromal biomarker. Impaired olfactory function is a common finding in PD patients, occurring early in the course of the disease [76] and may be able to distinguish PD from other movement disorders [77]. Similarly, studies have shown that RBD may precede the onset of parkinsonism [78] in addition to other prodromal symptoms of PD [79], which include depression, constipation, and cardiac dysfunction (the foundation of cardiac scan, see Table 1). DAT imaging has been especially useful towards developing such a biomarker. In a cohort of 361 asymptomatic relatives of patients with PD, idiopathic olfactory dysfunction was assessed and nigrostriatal DA neuron function was evaluated through SPECT DAT imaging. A total of 12.5% of the hyposmic first-degree relatives of patients with PD eventually developed PD and all had an abnormal baseline SPECT scan [80]. RBD patients similarly have shown reduced striatal DAT binding using 123I-IPT SPECT [69,81], and to a lesser extent FP-CIT SPECT [82], indicating that neuroimaging, particularly DAT imaging, is capable of detecting prodromal DA dysfunction in subjects who are at risk of PD but have not yet developed motor symptoms, as mentioned above. This is indeed confirmed in our recent investigation (Figure 1). Longitudinally monitoring patients that have these conditions and observing the differences between those that develop PD and controls may provide valuable insight into prodromal markers of PD in addition to markers capable of monitoring disease progression.
Figure 1. Imaging of dopamine transporter in different types of RBD patients.

(a) Idiopathic RBD with normal DAT imaging and no signs for PD (male, 66 yrs); (b) Idiopathic RBD with reduction of DAT in imaging but no signs for PD; however, PD was confirmed clinically after follow up with left limbs affected at onset (male, 70 yrs); (C) Idiopathic RBD with reduction of DAT in imaging and PD (male, 71 yrs).
Patients with LRRK2 or GBA mutations or with prodromal PD symptoms can also be used to develop biochemical biomarkers. Most of the work in this area has focused on LRRK2 carriers. Plotting PLS-DA scores calculated from asymptomatic LRRK2 carriers and family members without a mutant copy of LRRK2 showed that metabolomic profiles of these groups were distinguishable from each other [54]. Additionally, decreased levels of Aβ and tau species in CSF correlate with decreased DA neuron function in LRRK2 mutation carriers as detected by imaging through 18F-dopa, 11C-DTBZ and 11C-methylphenidate [83], adding strength to their usefulness as PD biomarkers. Because asymptomatic carriers represent a prodromal state of PD, such findings indicate metabolomics and protein levels may be useful in the development of prodromal biomarkers for PD. Unfortunately, attempts at using α-synuclein or DJ-1 in LRRK2 patients for the same purpose has not proved successful [84], warranting further investigation into candidate markers in at-risk populations. Longitudinal studies on such populations will likely allow for the development of a prodromal biomarker for PD.
Transition from CSF to peripheral fluids
Although CSF biomarkers have shown great potential for PD diagnosis, CSF is obtained through lumbar puncture, which is relatively invasive compared with obtaining other biofluids such as blood or saliva. Although some studies in plasma have yielded promising results (discussed above) [40,41], further validation is still necessary. One recent study identified α-synuclein and DJ-1 in saliva [85], opening the door for using saliva as a biofluid in which to develop a biomarker for PD. Future studies should aim to integrate neuroimaging techniques in addition to markers in biofluids to develop a marker that is not only specific for PD but also allows for preclinical diagnosis and monitoring of disease progression.
Concluding remarks
The current field of PD biomarkers focuses on using neuroimaging in addition to biochemical markers that can be measured in biofluids. Although a perfect biomarker has not been developed, significant progress has been made towards discovering molecular and imaging patterns that can accurately distinguish PD and monitor its progression. Among imaging methods that are currently available, DAT appears to be most widely used towards diagnosing PD, including defining PD at prodromal stages. However, newer imaging methods are needed to improve differential diagnosis of overlapping parkinsonian disorders and assessing PD progression objectively. Biochemical markers are important not only towards filling the gaps associated with imaging fields, but also in revealing novel molecular targets involved in PD development and progression. Additionally, biochemical markers are typically determined in human fluids, making them advantageous in the aspect that they are easily accessible for application in a routine clinical setting. Future efforts should look to integrate these two fields and extend studies to the prodromal stages of PD in addition to nonmotor symptoms. Correlating imaging data with biochemical markers will likely improve the diagnostic accuracy of PD, including at prodromal stages, in addition to monitoring its progression and the efficacy of treatment, especially at early phases when neuroprotective therapies are most effective.
Highlights.
Parkinson’s disease has appreciable rate of misdiagnosis.
Imaging and biochemical markers aim to assist with diagnosis and progression.
Imaging is more advanced, while biochemical markers may reflect pathogenesis.
Major areas of research are geared towards developing pre- or non-motor biomarkers.
Combining imaging and biochemical markers is likely to move the field effectively.
Acknowledgments
JW is supported by the grants (30600663, 81071018) from the Natural Science Foundation of China, Project (10PJ1401700) of Pujiang Talents at Shanghai, project (09411960900) from Science and Technology Commission of Shanghai Municipality, and project (2010-155) of Shanghai Municipal Health Bureau. JH is supported by a training grant from the NIH (T32 ES007032). CZ is supported by the grants (81171189) from the Natural Science Foundation of China. TC is supported by a training grant from NIEHS (T32 ES015459). JZ is supported by grants of MJFF as well as NIH grants [RO1-AG033398, P42- ES004696 (subaward 5897), P30-ES007033 (subaward 6364), RO1-ES016873, R01-ES019277, R01-NS057567, and P50-NS062684 (subaward 6221)].
Footnotes
Conflict of Interest: The authors report no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Braak H, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197–211. doi: 10.1016/s0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
- 2.Meara J, et al. Accuracy of diagnosis in patients with presumed Parkinson’s disease. Age Ageing. 1999;28:99–102. doi: 10.1093/ageing/28.2.99. [DOI] [PubMed] [Google Scholar]
- 3.Morrish PK, et al. Measuring the rate of progression and estimating the preclinical period of Parkinson’s disease with [18F]dopa PET. J Neurol Neurosurg Psychiatry. 1998;64:314–319. doi: 10.1136/jnnp.64.3.314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Fearnley JM, Lees AJ. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain. 1991;114:2283–2301. doi: 10.1093/brain/114.5.2283. [DOI] [PubMed] [Google Scholar]
- 5.Morrish PK, et al. Clinical and [18F] dopa PET findings in early Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1995;59:597–600. doi: 10.1136/jnnp.59.6.597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Brooks DJ, et al. The relationship between locomotor disability, autonomic dysfunction, and the integrity of the striatal dopaminergic system in patients with multiple system atrophy, pure autonomic failure, and Parkinson’s disease, studied with PET. Brain. 1990;113:1539–1552. doi: 10.1093/brain/113.5.1539. [DOI] [PubMed] [Google Scholar]
- 7.Vingerhoets FJ, et al. Which clinical sign of Parkinson’s disease best reflects the nigrostriatal lesion? Ann Neurol. 1997;41:58–64. doi: 10.1002/ana.410410111. [DOI] [PubMed] [Google Scholar]
- 8.Ribeiro MJ, et al. Dopaminergic function and dopamine transporter binding assessed with positron emission tomography in Parkinson disease. Arch Neurol. 2002;59:580–586. doi: 10.1001/archneur.59.4.580. [DOI] [PubMed] [Google Scholar]
- 9.Brooks DJ, Pavese N. Imaging biomarkers in Parkinson’s disease. Prog Neurobiol. 2011;95:614–628. doi: 10.1016/j.pneurobio.2011.08.009. [DOI] [PubMed] [Google Scholar]
- 10.Benamer TS, et al. Accurate differentiation of parkinsonism and essential tremor using visual assessment of [123I]-FP-CIT SPECT imaging: the [123I]-FP-CIT study group. Mov Disord. 2000;15:503–510. [PubMed] [Google Scholar]
- 11.Marshall VL, et al. Two-year follow-up in 150 consecutive cases with normal dopamine transporter imaging. Nucl Med Commun. 2006;27:933–937. doi: 10.1097/01.mnm.0000243374.11260.5b. [DOI] [PubMed] [Google Scholar]
- 12.Wang J, et al. 18F-FP-CIT PET imaging and SPM analysis of dopamine transporters in Parkinson’s disease in various Hoehn & Yahr stages. J Neurol. 2007;254:185–190. doi: 10.1007/s00415-006-0322-9. [DOI] [PubMed] [Google Scholar]
- 13.Fahn S, et al. Levodopa and the progression of Parkinson’s disease. N Engl J Med. 2004;351:2498–2508. doi: 10.1056/NEJMoa033447. [DOI] [PubMed] [Google Scholar]
- 14.Bohnen NI, et al. Positron emission tomography of monoaminergic vesicular binding in aging and Parkinson disease. J Cereb Blood Flow Metab. 2006;26:1198–1212. doi: 10.1038/sj.jcbfm.9600276. [DOI] [PubMed] [Google Scholar]
- 15.Lee CS, et al. In vivo positron emission tomographic evidence for compensatory changes in presynaptic dopaminergic nerve terminals in Parkinson’s disease. Ann Neurol. 2000;47:493–503. [PubMed] [Google Scholar]
- 16.Okamura N, et al. In vivo measurement of vesicular monoamine transporter type 2 density in Parkinson disease with (18)F-AV-133. J Nucl Med. 2010;51:223–228. doi: 10.2967/jnumed.109.070094. [DOI] [PubMed] [Google Scholar]
- 17.Pavese N. Do we still need to develop new imaging biomarkers of dopamine denervation for Parkinson’s disease? Exp Neurol. 2011;233:118–120. doi: 10.1016/j.expneurol.2011.10.008. [DOI] [PubMed] [Google Scholar]
- 18.Jennings DL, et al. (123I) beta-CIT and single-photon emission computed tomographic imaging vs clinical evaluation in Parkinsonian syndrome: unmasking an early diagnosis. Arch Neurol. 2004;61:1224–1229. doi: 10.1001/archneur.61.8.1224. [DOI] [PubMed] [Google Scholar]
- 19.Masilamoni G, et al. (18)F-FECNT: validation as PET dopamine transporter ligand in parkinsonism. Exp Neurol. 2010;226:265–273. doi: 10.1016/j.expneurol.2010.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rinne JO, et al. PET study on striatal dopamine D2 receptor changes during the progression of early Parkinson’s disease. Mov Disord. 1993;8:134–138. doi: 10.1002/mds.870080203. [DOI] [PubMed] [Google Scholar]
- 21.Plotkin M, et al. Combined 123I-FP-CIT and 123I-IBZM SPECT for the diagnosis of parkinsonian syndromes: study on 72 patients. J Neural Transm. 2005;112:677–692. doi: 10.1007/s00702-004-0208-x. [DOI] [PubMed] [Google Scholar]
- 22.Antonini A, et al. Complementary PET studies of striatal neuronal function in the differential diagnosis between multiple system atrophy and Parkinson’s disease. Brain. 1997;120:2187–2195. doi: 10.1093/brain/120.12.2187. [DOI] [PubMed] [Google Scholar]
- 23.Eidelberg D, et al. The metabolic topography of parkinsonism. J Cereb Blood Flow Metab. 1994;14:783–801. doi: 10.1038/jcbfm.1994.99. [DOI] [PubMed] [Google Scholar]
- 24.Tang CC, et al. Differential diagnosis of parkinsonism: a metabolic imaging study using pattern analysis. Lancet Neurol. 2010;9:149–158. doi: 10.1016/S1474-4422(10)70002-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tang CC, et al. Abnormalities in metabolic network activity precede the onset of motor symptoms in Parkinson’s disease. J Neurosci. 2010;30:1049–1056. doi: 10.1523/JNEUROSCI.4188-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ma Y, et al. Abnormal metabolic network activity in Parkinson’s disease: test-retest reproducibility. J Cereb Blood Flow Metab. 2007;27:597–605. doi: 10.1038/sj.jcbfm.9600358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Asanuma K, et al. Network modulation in the treatment of Parkinson’s disease. Brain. 2006;129:2667–2678. doi: 10.1093/brain/awl162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Feigin A, et al. Modulation of metabolic brain networks after subthalamic gene therapy for Parkinson’s disease. Proc Natl Acad Sci USA. 2007;104:19559–19564. doi: 10.1073/pnas.0706006104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hilker R, et al. Dementia in Parkinson disease: functional imaging of cholinergic and dopaminergic pathways. Neurology. 2005;65:1716–1722. doi: 10.1212/01.wnl.0000191154.78131.f6. [DOI] [PubMed] [Google Scholar]
- 30.Martin WR, et al. Midbrain iron content in early Parkinson disease: a potential biomarker of disease status. Neurology. 2008;70:1411–1417. doi: 10.1212/01.wnl.0000286384.31050.b5. [DOI] [PubMed] [Google Scholar]
- 31.Scherfler C, et al. Voxel-wise analysis of diffusion weighted imaging reveals disruption of the olfactory tract in Parkinson’s disease. Brain. 2006;129:538–542. doi: 10.1093/brain/awh674. [DOI] [PubMed] [Google Scholar]
- 32.Vaillancourt DE, et al. High-resolution diffusion tensor imaging in the substantia nigra of de novo Parkinson disease. Neurology. 2009;72:1378–1384. doi: 10.1212/01.wnl.0000340982.01727.6e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chan LL, et al. Case control study of diffusion tensor imaging in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2007;78:1383–1386. doi: 10.1136/jnnp.2007.121525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Polymeropoulos MH, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–2047. doi: 10.1126/science.276.5321.2045. [DOI] [PubMed] [Google Scholar]
- 35.Ahn TB, et al. alpha-Synuclein gene duplication is present in sporadic Parkinson disease. Neurology. 2008;70:43–49. doi: 10.1212/01.wnl.0000271080.53272.c7. [DOI] [PubMed] [Google Scholar]
- 36.Tokuda T, et al. Decreased alpha-synuclein in cerebrospinal fluid of aged individuals and subjects with Parkinson’s disease. Biochem Biophys Res Commun. 2006;349:162–166. doi: 10.1016/j.bbrc.2006.08.024. [DOI] [PubMed] [Google Scholar]
- 37.Mollenhauer B, et al. Direct quantification of CSF alpha-synuclein by ELISA and first cross-sectional study in patients with neurodegeneration. Exp Neurol. 2008;213:315–325. doi: 10.1016/j.expneurol.2008.06.004. [DOI] [PubMed] [Google Scholar]
- 38.Hong Z, et al. DJ-1 and alpha-synuclein in human cerebrospinal fluid as biomarkers of Parkinson’s disease. Brain. 2010;133:713–726. doi: 10.1093/brain/awq008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mollenhauer B, et al. α-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: a cohort study. Lancet Neurol. 2011;10:230–240. doi: 10.1016/S1474-4422(11)70014-X. [DOI] [PubMed] [Google Scholar]
- 40.Tokuda T, et al. Detection of elevated levels of {alpha}-synuclein oligomers in CSF from patients with Parkinson disease. Neurology. 2010;75:1766–1770. doi: 10.1212/WNL.0b013e3181fd613b. [DOI] [PubMed] [Google Scholar]
- 41.El-Agnaf OM, et al. Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006;20:419–425. doi: 10.1096/fj.03-1449com. [DOI] [PubMed] [Google Scholar]
- 42.Wang Y, et al. Phosphorylated α-Synuclein in Parkinson’s Disease. Sci Transl Med. 2012;4:121ra120. doi: 10.1126/scitranslmed.3002566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Waragai M, et al. Increased level of DJ-1 in the cerebrospinal fluids of sporadic Parkinson’s disease. Biochem Biophys Res Commun. 2006;345:967–972. doi: 10.1016/j.bbrc.2006.05.011. [DOI] [PubMed] [Google Scholar]
- 44.Waragai M, et al. Plasma levels of DJ-1 as a possible marker for progression of sporadic Parkinson’s disease. Neurosci Lett. 2007;425:18–22. doi: 10.1016/j.neulet.2007.08.010. [DOI] [PubMed] [Google Scholar]
- 45.Shi M, et al. Significance and confounders of peripheral DJ-1 and alpha-synuclein in Parkinson’s disease. Neurosci Lett. 2010;480:78–82. doi: 10.1016/j.neulet.2010.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pankratz N, et al. Genomewide association study for susceptibility genes contributing to familial Parkinson disease. Hum Genet. 2009;124:593–605. doi: 10.1007/s00439-008-0582-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Satake W, et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson’s disease. Nat Genet. 2009;41:1303–1307. doi: 10.1038/ng.485. [DOI] [PubMed] [Google Scholar]
- 48.Montine TJ, et al. CSF Aβ(42) and tau in Parkinson’s disease with cognitive impairment. Mov Disord. 2010;25:2682–2685. doi: 10.1002/mds.23287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Shi M, et al. Cerebrospinal fluid biomarkers for Parkinson disease diagnosis and progression. Ann Neurol. 2011;69:570–580. doi: 10.1002/ana.22311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Alves G, et al. CSF amyloid-beta and tau proteins, and cognitive performance, in early and untreated Parkinson’s disease: the Norwegian ParkWest study. J Neurol Neurosurg Psychiatry. 2010;81:1080–1086. doi: 10.1136/jnnp.2009.199950. [DOI] [PubMed] [Google Scholar]
- 51.Buongiorno M, et al. Amyloid-β and τ biomarkers in Parkinson’s disease-dementia. J Neurol Sci. 2011;310:25–30. doi: 10.1016/j.jns.2011.06.046. [DOI] [PubMed] [Google Scholar]
- 52.Bogdanov M, et al. Metabolomic profiling to develop blood biomarkers for Parkinson’s disease. Brain. 2008;131:389–396. doi: 10.1093/brain/awm304. [DOI] [PubMed] [Google Scholar]
- 53.Maetzler W, et al. Reduced but not oxidized cerebrospinal fluid glutathione levels are lowered in Lewy body diseases. Mov Disord. 2011;26:176–181. doi: 10.1002/mds.23358. [DOI] [PubMed] [Google Scholar]
- 54.Johansen KK, et al. Metabolomic profiling in LRRK2-related Parkinson’s disease. PLoS One. 2009;4:E7551. doi: 10.1371/journal.pone.0007551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ascherio A, et al. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol. 2009;66:1460–1468. doi: 10.1001/archneurol.2009.247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sun CC, et al. Association of serum uric acid levels with the progression of Parkinson’s disease in Chinese patients. Chin Med J (Engl) 2012;125:583–587. [PubMed] [Google Scholar]
- 57.Ahmed SS, et al. Metabolic profiling of Parkinson’s disease: evidence of biomarker from gene expression analysis and rapid neural network detection. J Biomed Sci. 2009;16:63. doi: 10.1186/1423-0127-16-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Scherzer CR, et al. Molecular markers of early Parkinson’s disease based on gene expression in blood. Proc Natl Acad Sci USA. 2007;104:955–960. doi: 10.1073/pnas.0610204104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Shadrina MI, et al. Expression analysis of suppression of tumorigenicity 13 gene in patients with Parkinson’s disease. Neurosci Lett. 2010;473:257–259. doi: 10.1016/j.neulet.2010.02.061. [DOI] [PubMed] [Google Scholar]
- 60.Filatova EV, et al. Expression of GSK3B gene in peripheral blood in patient with Parkinson’s disease. Mol Biol. 2011;45:459–463. [PubMed] [Google Scholar]
- 61.Grünblatt E, et al. Pilot study: peripheral biomarkers for diagnosing sporadic Parkinson’s disease. J Neural Transm. 2010;117:1387–1393. doi: 10.1007/s00702-010-0509-1. [DOI] [PubMed] [Google Scholar]
- 62.Molochnikov L, et al. A molecular signature in blood identifies early Parkinson’s disease. Mol Neurodegener. 2012;7:26. doi: 10.1186/1750-1326-7-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Abdi F, et al. Detection of biomarkers with a multiplex quantitative proteomic platform in cerebrospinal fluid of patients with neurodegenerative disorders. J Alzheimers Dis. 2006;9:293–348. doi: 10.3233/jad-2006-9309. [DOI] [PubMed] [Google Scholar]
- 64.Zhang J, et al. CSF multianalyte profile distinguishes Alzheimer and Parkinson diseases. Am J Clin Pathol. 2008;129:526–529. doi: 10.1309/W01Y0B808EMEH12L. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sinha A, et al. Identification of differentially displayed proteins in cerebrospinal fluid of Parkinson’s disease patients: a proteomic approach. Clin Chim Acta. 2009;400:14–20. doi: 10.1016/j.cca.2008.09.026. [DOI] [PubMed] [Google Scholar]
- 66.Wang ES, et al. Tetranectin and apolipoprotein A-I in cerebrospinal fluid as potential biomarkers for Parkinson’s disease. Acta Neurol Scand. 2010;122:350–359. doi: 10.1111/j.1600-0404.2009.01318.x. [DOI] [PubMed] [Google Scholar]
- 67.Berg D, et al. Echogenicity of the substantia nigra in Parkinson’s disease and its relation to clinical findings. J Neurol. 2001;248:684–689. doi: 10.1007/s004150170114. [DOI] [PubMed] [Google Scholar]
- 68.Gaenslen A, et al. The specificity and sensitivity of transcranial ultrasound in the differential diagnosis of Parkinson’s disease: a prospective blinded study. Lancet Neurol. 2008;7:417–424. doi: 10.1016/S1474-4422(08)70067-X. [DOI] [PubMed] [Google Scholar]
- 69.Iranzo A, et al. Decreased striatal dopamine transporter uptake and substantia nigra hyperechogenicity as risk markers of synucleinopathy in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study [corrected] Lancet Neurol. 2010;9:1070–1077. doi: 10.1016/S1474-4422(10)70216-7. [DOI] [PubMed] [Google Scholar]
- 70.Berg D, et al. Enlarged substantia nigra hyperechogenicity and risk for Parkinson disease: a 37-month 3-center study of 1847 older persons. Arch Neurol. 2011;68:932–937. doi: 10.1001/archneurol.2011.141. [DOI] [PubMed] [Google Scholar]
- 71.Berg D, et al. Five-year follow-up study of hyperechogenicity of the substantia nigra in Parkinson’s disease. Mov Disord. 2005;20:383–385. doi: 10.1002/mds.20311. [DOI] [PubMed] [Google Scholar]
- 72.Healy DG, et al. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: a case-control study. Lancet Neurol. 2008;7:583–590. doi: 10.1016/S1474-4422(08)70117-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Adams JR, et al. PET in LRRK2 mutations: comparison to sporadic Parkinson’s disease and evidence for presymptomatic compensation. Brain. 2005;128:2777–2785. doi: 10.1093/brain/awh607. [DOI] [PubMed] [Google Scholar]
- 74.Nandhagopal R, et al. Progression of dopaminergic dysfunction in a LRRK2 kindred: a multitracer PET study. Neurology. 2008;71:1790–1795. doi: 10.1212/01.wnl.0000335973.66333.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kono S, et al. Functional brain imaging in glucocerebrosidase mutation carriers with and without parkinsonism. Mov Disord. 2010;25:1823–1829. doi: 10.1002/mds.23213. [DOI] [PubMed] [Google Scholar]
- 76.Doty RL, et al. Bilateral olfactory dysfunction in early stage treated and untreated idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1992;55:138–142. doi: 10.1136/jnnp.55.2.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Doty RL, et al. Olfactory testing differentiates between progressive supranuclear palsy and idiopathic Parkinson’s disease. Neurology. 1993;43:962–965. doi: 10.1212/wnl.43.5.962. [DOI] [PubMed] [Google Scholar]
- 78.Schenck CH, et al. Delayed emergence of a parkinsonian disorder in 38% of 29 older men initially diagnosed with idiopathic rapid eye movement sleep behaviour disorder. Neurology. 1996;46:388–393. doi: 10.1212/wnl.46.2.388. [DOI] [PubMed] [Google Scholar]
- 79.Fantini ML, et al. Olfactory deficit in idiopathic rapid eye movements sleep behavior disorder. Brain Res Bull. 2006;70:386–390. doi: 10.1016/j.brainresbull.2006.07.008. [DOI] [PubMed] [Google Scholar]
- 80.Ponsen MM, et al. Olfactory testing combined with dopamine transporter imaging as a method to detect prodromal Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81:396–399. doi: 10.1136/jnnp.2009.183715. [DOI] [PubMed] [Google Scholar]
- 81.Eisensehr I, et al. Reduced striatal dopamine transporters in idiopathic rapid eye movement sleep behaviour disorder. Comparison with Parkinson’s disease and controls. Brain. 2000;123:1155–1160. doi: 10.1093/brain/123.6.1155. [DOI] [PubMed] [Google Scholar]
- 82.Stiasny-Kolster K, et al. Combination of ‘idiopathic’ REM sleep behaviour disorder and olfactory dysfunction as possible indicator for alpha-synucleinopathy demonstrated by dopamine transporter FP-CIT-SPECT. Brain. 2005;128:126–137. doi: 10.1093/brain/awh322. [DOI] [PubMed] [Google Scholar]
- 83.Aasly JO, et al. Cerebrospinal fluid amyloid β and tau in LRRK2 mutation carriers. Neurology. 2012;78:55–61. doi: 10.1212/WNL.0b013e31823ed101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Shi M, et al. DJ-1 and αSYN in LRRK2 CSF do not correlate with striatal dopaminergic function. Neurobiol Aging. 2012;33:836, E835–E837. doi: 10.1016/j.neurobiolaging.2011.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Devic I, et al. Salivary alpha-synuclein and DJ-1: potential biomarkers for Parkinson’s disease. Brain. 2011;134:E178. doi: 10.1093/brain/awr015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Madras BK, et al. Altropane, a SPECT or PET imaging probe for dopamine neurons: III. Human dopamine transporter in postmortem normal and Parkinson’s diseased brain. Synapse. 1998;29:116–127. doi: 10.1002/(SICI)1098-2396(199806)29:2<116::AID-SYN3>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
- 87.Eckert T, et al. FDG PET in the differential diagnosis of parkinsonian disorders. Neuroimage. 2005;26:912–921. doi: 10.1016/j.neuroimage.2005.03.012. [DOI] [PubMed] [Google Scholar]
