Skip to main content
Brain logoLink to Brain
. 2023 Aug 3;146(12):4845–4869. doi: 10.1093/brain/awad265

How should we be using biomarkers in trials of disease modification in Parkinson’s disease?

Nirosen Vijiaratnam 1, Thomas Foltynie 2,
PMCID: PMC10690028  PMID: 37536279

Abstract

The recent validation of the α-synuclein seed amplification assay as a biomarker with high sensitivity and specificity for the diagnosis of Parkinson’s disease has formed the backbone for a proposed staging system for incorporation in Parkinson’s disease clinical studies and trials. The routine use of this biomarker should greatly aid in the accuracy of diagnosis during recruitment of Parkinson’s disease patients into trials (as distinct from patients with non-Parkinson’s disease parkinsonism or non-Parkinson’s disease tremors). There remain, however, further challenges in the pursuit of biomarkers for clinical trials of disease modifying agents in Parkinson’s disease, namely: optimizing the distinction between different α-synucleinopathies; the selection of subgroups most likely to benefit from a candidate disease modifying agent; a sensitive means of confirming target engagement; and the early prediction of longer-term clinical benefit. For example, levels of CSF proteins such as the lysosomal enzyme β-glucocerebrosidase may assist in prognostication or allow enrichment of appropriate patients into disease modifying trials of agents with this enzyme as the target; the presence of coexisting Alzheimer’s disease-like pathology (detectable through CSF levels of amyloid-β42 and tau) can predict subsequent cognitive decline; imaging techniques such as free-water or neuromelanin MRI may objectively track decline in Parkinson’s disease even in its later stages. The exploitation of additional biomarkers to the α-synuclein seed amplification assay will, therefore, greatly add to our ability to plan trials and assess the disease modifying properties of interventions. The choice of which biomarker(s) to use in the context of disease modifying clinical trials will depend on the intervention, the stage (at risk, premotor, motor, complex) of the population recruited and the aims of the trial. The progress already made lends hope that panels of fluid biomarkers in tandem with structural or functional imaging may provide sensitive and objective methods of confirming that an intervention is modifying a key pathophysiological process of Parkinson’s disease. However, correlation with clinical progression does not necessarily equate to causation, and the ongoing validation of quantitative biomarkers will depend on insightful clinical-genetic-pathophysiological comparisons incorporating longitudinal biomarker changes from those at genetic risk with evidence of onset of the pathophysiology and those at each stage of manifest clinical Parkinson’s disease.

Keywords: Parkinson’s disease, biomarkers, disease modification, clinical trials


Vijiaratnam and Foltynie consider how fluid and imaging biomarkers can best be used in Parkinson’s disease trials to help select participants most likely to benefit from candidate disease-modifying agents, confirm target engagement, and track disease progression and treatment response.

Introduction

Modifying the relentless deteriorating course of Parkinson’s disease (PD) remains a critical yet currently elusive goal. Despite decades of trials evaluating promising candidates, no treatments have yet been proven to achieve this. While this may be due to lack of trial evaluation of truly effective agents, other potentially contributing factors include imprecise patient selection, inadequacies in trial design, failure to confirm target engagement and the absence of objective measures of disease progression.1

One way of improving the likelihood of success is by identifying better biomarkers. A biomarker is a characteristic that is objectively measured and evaluated from any substance, structure or process that can be measured in the body or its products as an indicator of normal biological or pathogenic processes, or pharmacologic responses to a therapeutic intervention.2 An ideal biomarker should be readily quantifiable in accessible clinical samples [clinical assessments, biofluids (blood, CSF, urine, saliva, tears, stool), imaging and tissues (skin, oro-gastrointestinal mucosa)], while being reliable, quick and inexpensive.

Suboptimal patient selection in disease modifying trials may be related to poor diagnostic accuracy. Pathological modification (phosphorylation and conformational transformation) of the physiological protein α-synuclein (α-syn) to misfolded oligomeric and fibrillary forms is the most consistent pathological feature of PD.3 The accumulation and interplay of these abnormal protein forms with the organelles/cellular pathways involved in their clearance as well as normal cellular maintenance and survival results in neuronal dysfunction and ultimately axonal injury and neuronal death.

The α-syn seed amplification assay (SAA) has high sensitivity and specificity for PD diagnostic accuracy, with a recent study of >1100 samples from the Parkinson’s progression markers initiative (PPMI) cohort4 further confirming pre-existing evidence for its use,5–11 and is now proposed as a core aspect of a potential staging system for PD.12,13 This is potentially a pivotal step in clarifying eligibility criteria for inclusion in trials and distinguishing PD patients from those with atypical forms of parkinsonism. While needing further clarification, the α-syn SAA is at the present time largely a binary measure simply indicating the presence/absence of the pathophysiological process of α-syn aggregation and cannot yet be used to track disease severity, which instead relies on clinical measurements.

As such, there is still a need for additional biomarkers that might enrich treatment arms for PD subgroups most likely to respond and allow early exploratory analyses according to engagement of the therapeutic with its putative target. Current trials typically rely on clinical end points with scales and questionnaires, which are subject to inter-rater variability, while potentially being confounded by symptomatic drug effects. Evaluations using scales may also be compromised by non-linear changes over time,14 be limited by reduced compliance, recall bias and fatigue,15 sometimes do not correlate sufficiently with quantitative objective assessments16,17 and vary in their sensitivity at different disease stages,18,19 raising questions about the inclusion of patients who may have progressed beyond the salvageable period.

Biomarkers that are robustly demonstrated to track disease progression and treatment effects could potentially shorten periods of assessment and reduce the number of patients required for preliminary demonstration of efficacy. Ideally, short-term changes in the biomarker should anticipate long-term clinical outcomes. Furthermore, by confirming target engagement according to the dose(s) of the agent under study, biomarkers can be used to improve the distinction between an intervention’s disease-modifying effects and purely symptomatic improvements. While there are parallel efforts exploring additional biomarkers for PD prior to clinically manifest disease, in this review, we will discuss the current state of fluid, tissue and imaging biomarker developments in clinically established PD and their potential for use either alone or in combination in future disease modifying clinical trials.

Fluid and tissue biomarkers

Box 1 outlines techniques that have been used to measure different forms of α-syn as well as other protein/enzyme levels that reflect cellular pathway abnormalities that can be measured in biofluids.

Box 1. Fluid and tissue biomarker measurement techniques.

ELISA

  • target-specific antibodies bind to the sample proteins

  • secondary antibody linked to an enzyme recognizes the matched antibodies

  • fluorescent reaction is created when exposed to a chemical substrate

  • amount of antigen present correlates to intensity of colour change

  • detection range inferior to other high-sensitivity techniques

Luminex

  • beads conjugated with antibody against specific analyte present different colour codes

  • high-throughput screening

  • can measure up to 80 different proteins or RNA from a single microplate

Mesoscale discovery

  • high-throughput measurement of single or multiple targets

  • antibodies can be conjugated to generate electro chemiluminescent signals, unlike ELISA

Single molecule array

  • antibody-based ELISA and bead-based platform

  • antibody-coated bead binds to a single molecule and analysed separately

  • multiplexing of up to 11 analytes, high sensitivity and wide detection range

Proximity extension assay

  • DNA oligonucleotide tags linked to matched antibodies that both bind to target protein

  • antibodies come into proximity on binding, DNA duplex formed, sequence amplified

  • wide library of matched antibodies with high sensitivity and specificity for their targets

SomaScan

  • Aptamers (short, single-stranded DNA or RNA molecules) bind target

  • quantified by microarrays or quantitative PCR

  • allows creation of library with high sensitivity for targets

Single molecule counting

  • antibody–antigen sandwich complexes from either beads or plates

  • broken up and fluorescently labelled detection antibody counted by laser beam

  • allows for a high dynamic concentration range

Mass spectrometry

  • measures mass-to-charge ratio of one or more molecules present

  • provide quantitative information about composition of complex protein samples

  • can also provide information about conformational properties

Microscopy

  • used to examine to structure and formation of aggregates

  • approaches include fluorescence (aggregates labelled with fluorescent probes) microscopy and electron microscopy (resolve oligomer structure at higher resolution)

Seed amplification assays

  • aggregation assays that detect the presence of protein aggregates

  • sample sonication and incubation with recombinant protein monomer

  • aggregate seeds template and induce aggregation of the excess protein monomers

  • reaction monitored by a thioflavin readout, aggregation curve characteristics recorded

Extracellular vesicles protein measurement

  • released by cells, content represent central nervous system processes

  • precipitation to increase concentration and neuronal enrichment with immune capture

  • protein quantification with electrochemiluminescence (e.g. mesoscale discovery)

Alpha-synuclein

Total, phosphorylated and oligomeric α-syn levels and their ratios in CSF, blood and other body fluids and tissues have all been explored for biomarker use (Table 1).

Table 1.

Alpha-synuclein fluid and tissue biomarkers and their potential relevance to clinical trial design

Biomarker Origin Differentiating PD from healthy controls Marker of disease severity Differentiating PD from atypical parkinsonism Predicting disease progression Surrogate for disease progression
Total α-syn CSF + +
CSF (exosomes) + +
Plasma/serum ++ +
Plasma/serum (exosomes) +++ ++ ++ + +
Saliva +++ +
Tears ++
Skin ++ +
Ser-129p-α-syn CSF ++ +++ ++ +
Serum/plasma + + +
Tissue/intestine ++
Skin + ++
Ratio of phosphorylated α-syn to total α-syn CSF +
Saliva (exosomes) +
Tyrosine phosphorylated α-syn CSF +
Tyrosine nitrated α-syn Serum +
Oligomeric α-syn CSF +++ ++ +
Plasma/serum/blood +
Serum/plasma (exosomes) + + +
Saliva +++ +
Saliva (exosomes) +
Ratio of oligomeric to total α-syn CSF +++ + + + +
Plasma/serum (exosomes) +
Saliva +
Red blood cells +
Oligomeric phosphorylated α-syn species CSF +
Plasma +
α-syn seed amplification CSF +++ + +++
Saliva +
GI biopsy +
Skin ++
Olfactory mucosa + +

α-syn = α-synuclein; GI = gastrointestinal tract; PD = Parkinson’s disease. Grading approach adapted from Majbour et al.20

− = No effect (also scored if negative in a meta-analysis).

+ = Effect in 1 study/inconsistent results across studies.

++ = Effect in 2–3 studies using single site cohort.

+++ = Effect in ≥3 studies or multisite cohort (also scored if positive in meta-analysis).

Distinguishing Parkinson’s disease from other conditions

Total free α-syn levels have been explored in CSF, plasma/serum, saliva and submandibular gland tissue and are of no diagnostic value in PD.21–30 Measurement of total α-syn levels in extracellular vesicles (EVs) either in CSF,31 plasma/serum32–39 or saliva40 can distinguish PD from controls.33–37,39,41–43 Total α-syn levels in EVs derived from neurons can also distinguish PD from atypical disorders, though best distinction is achieved when α-syn levels are combined with levels of other proteins such as clusterin.36,44 Similarly, differences in α-syn levels in neuronal- compared with oligodendroglial-derived EVs shows promise for distinguishing PD from multiple system atrophy (MSA).38 Phosphorylated α-syn at serine-129 (Ser-129p-α-syn) levels are elevated in the CSF,25,45–48 serum and plasma49–52 of PD patients, although similar elevations are seen in atypical parkinsonian conditions, limiting specificity/diagnostic use.53–56 Elevated levels are similarly seen for Ser-129p-α-syn in skin.30,57–61 A predilection for Ser-129p-α-syn deposition in autonomic compared with somatosensory nerve fibres and proximal to distal gradients could be applied for improving the distinction between PD and MSA-parkinsonian type (MSA-P).62,63

Levels of α-syn oligomers are also increased in the CSF,28,48,64–68 plasma,69,70 red blood cells,71,72 saliva and tears29,64,73–78 of PD patients (although again with a few teams reporting contradictory findings67,79,80). Oligomeric CSF α-syn levels taken alone, however, have unsatisfactory diagnostic properties.25 Combining oligomeric α-syn and aggregated tau measurement in serum neuronal-derived exosomes seems to distinguish PD from tauopathies well.81 Reliable quantification and differentiation approaches between protein species (oligomers, fibrils and other aggregated forms) are currently lacking.51,53 Making these distinctions will be critical in improving the diagnostic performance of aggregated forms, considering unique patterns have been noted in different synucleinopathies.82,83 Ratios of Ser-129p-α-syn and/or oligomeric α-syn to total α-syn are elevated in PD and seem most promising in overcoming the limitations of individual markers for differentiating synucleinopathies.45,46,54,55,66,68,84,85

Seed amplification assays such as real-time quaking-induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA) are arguably the most important achievement in the field of biomarkers to date and will likely be the most useful diagnostic biomarker for trials. These techniques can amplify and detect minute amounts of aggregated α-syn in CSF.10,86–88 Studies comparing brain and CSF samples have demonstrated excellent performance for distinguishing PD from healthy controls (sensitivity and specificity: 90–100%),4–11 with comparable results for both seeding methods7,10 across laboratories.10 Assays can also distinguish PD from non-synuclein disorders such as progressive supranuclear palsy (PSP) and corticobasal syndromes (CBS),11 although the accuracy in distinguishing MSA from these conditions is poor (sensitivity: 4–82%), while studies exploring the use of α-syn SAA to distinguish MSA from PD have also reported variable findings.87–91 As differences in α-syn strains and therefore biochemical, morphological and structural properties of the final α-syn SAA reaction products underlie PD and MSA phenotypic heterogeneity, different outcomes may be explained by the fact that different chemical environments (SAA reaction mixes) can differentially influence the formation and growth of different strains. Protocols optimized for PD may not therefore work so well for MSA detection.11,92

In attempts to avoid lumbar puncture, the use of α-syn SAA has been explored using samples obtained through less invasive approaches. Increased α-syn skin seeding activity has been observed in PD (post-mortem and living) patients with excellent distinction from non-neurodegenerative cases,93 while aggregation rates using RT QuIC correlate with cognitive and motor status.8 Similarly, seeding activity in submandibular gland tissue of PD patients has been noted, although sensitivity (73.2% versus 100%) and specificity (78.6% versus 94%) for distinguishing PD from healthy controls varies between studies,94,95 while preliminary findings in saliva are promising.96 A recent report demonstrating the excellent ability of serum immunoprecipitation-based RT-QuIC for distinguishing PD from healthy controls may herald a new approach to diagnosing PD via a simple blood test, although lower detection rates in MSA, likely due to technical factors, will still need to be overcome.97 Similarly, the demonstration of seeding activity from pathological α-syn derived from plasma EVs is also promising.98 The use of less invasive samples will be ideal for trial recruitment (given feedback from patients regarding tolerability of submandibular gland biopsy) but will require demonstration of comparability with the high sensitivity and specificity achieved with CSF (although a recent meta-analysis suggested comparability between CSF and skin for diagnostic purposes90,99).

Predicting severity phenotypes and measuring progression

Total free α-syn levels do not correlate with disease severity and their ability to predict and track progression is also poor.22,25,49 EV total α-syn levels also predict and track progression in PD poorly.31,32,35,36,100

While Ser-129p-α-syn levels do seem to reflect disease severity45,46,101 and motor symptom progression,102 an inverse relationship in later disease (potentially as a result of extensive neuronal damage)53,103 makes its use as a progression biomarker challenging if applied to trials with long-term follow-up or involving patients with established disease. CSF and serum levels of a number of other phosphorylated α-syn species have also been explored, although preliminary findings are somewhat conflicting.104–106 A rostro-caudal pSer129-α-syn deposition gradient in the gastrointestinal tract of PD patients has also been noted, reflecting neurodegeneration in the myenteric plexus,107,108 although this may be a reactive physiological phenomenon.109 Disentangling reactive from pathological components will be important, as deposition may occur here earlier and therefore guide earlier treatment in early motor stages where diagnostic criteria have yet to be totally fulfilled.

Oligomeric CSF α-syn levels can also reflect PD severity and progression,47,54,101,103 despite some contradictory evidence,110 although previously highlighted limitations of differentiating aggregated forms need to be addressed. Longitudinal measurement of Ser-129p-α-syn and/or oligomeric to total α-syn ratios might detect effective treatment responses.45,46,54,66,68,84,101 Similar findings have also been observed when measuring these ratios in serum and salivary EVs, although this does not seem to offer additional value.35,36,39,85,111,112

Correlation of α-syn SAA with disease severity and progression is unclear, and specific kinetic cut-offs remain elusive, although quantification of α-syn SAA end products with oligomer-specific enzyme-linked immunosorbent assay (ELISA) may be helpful in this regard.10,20,113 Taken together, the best α-syn candidate biomarkers for diagnosing PD to consider for clinical trials are α-syn SAA. The ratios of Ser-129p-α-syn and or oligomeric α-syn to total α-syn can also helpfully differentiate between synucleinopathies45,46,54,66,68,84 and are credible markers for tracking progression.

Alzheimer’s disease-like biomarkers

Amyloid-β (Aβ) peptides are cleaved from the amyloid precursor protein (APP) into the peptides Aβ42 and Aβ40, which can form extracellular amyloid plaques.114,115 Tau proteins comprise highly soluble isoforms, while their hyperphosphorylation contributes to the development of neurofibrillary tangles (NFTs).116 Amyloid plaques are abundant in the CNS alongside NFTs in Alzheimer’s disease (AD), while NFTs are characteristic of PSP and CBS.117,118

Distinguishing Parkinson’s disease from other conditions

Biomarkers reflecting tau and amyloid pathology can be measured in CSF and blood and include free and EV levels of total tau (t-tau), phosphorylated tau (p-tau) and amyloid peptide isoforms (Aβ42 and Aβ40). Higher CSF t-tau and decreased Aβ42 levels occur in tauopathies. This combination best distinguishes PD from CBS, although the relative rarity of CBS makes widespread testing in PD trials of modest value.119,120 Preliminary evidence suggests that ultrasensitive tau SAA may identify/exclude patients with tauopathies from PD at trial recruitment,121 although a combined assay with α-syn would be more ideal.

The combination of reduced Aβ42 and increased t-tau and p-tau levels is collectively termed ‘an AD-like profile’ considering its specificity for diagnosing the condition.122 This profile occurs in a larger proportion of synucleinopathy patients with prominent cognitive dysfunction, i.e. Parkinson’s disease dementia (PDD) and dementia with Lewy bodies (DLB).123–125 CSF AD-like biomarkers may, therefore, be useful for differentiating DLB from other parkinsonian disorders, although for some interventional trials this distinction may be somewhat arbitrary. Levels of t-tau and p-tau are increased in all parkinsonian disease groups and combining them with Aβ42 only usefully differentiates PD from frontotemporal dementia.126 Taken together, these findings suggest free blood levels of these markers are unlikely to be of diagnostic value in trials.

Predicting severity phenotypes and measuring progression

Tau and AD pathology commonly coexist in synucleinopathy patients127 and correlate with an acceleration in cognitive decline.128,129 PD patients with lower CSF Aβ42 levels at disease onset also have earlier appearance of cognitive impairment and more rapid conversion to PD-related dementia.68,130,131 The measurement of CSF Aβ42 could, therefore, be of prognostic value by reflecting brain amyloid content even prior to apparent clinical cognitive impairment.132

Although Aβ42 and tau can also be measured in blood, levels correlate poorly with cerebral pathology,133 potentially due to extra-cerebral sources such as platelets. Ultrasensitive immunoassay technologies such as immunomagnetic reduction (IMR) improve this,134 although correlation with cognitive function has been inconsistent.126,135,136 Similarly, total tau protein blood findings have been variable,135,136 potentially due to rapid changes in blood concentrations,137 although higher t-tau levels seem to correlate with lower cognitive performance.138

42 and tau can also be detected in EVs. While also not of diagnostic value, elevated levels in combination with elevated α-syn139,140 and lower serine phosphorylated insulin receptor substrate (IRS-p312), a marker of neuronal insulin resistance in blood EVs,141 predict worse motor and cognitive dysfunction progression phenotypes well. Larger replication studies of Aβ and tau in EVs are needed to better assess their validity for predicting cognitive dysfunction in PD before adoption for widespread use.

Measurement of other phosphorylated tau species (p-tau181, p-tau217 and p-tau231) in CSF and plasma can discriminate AD patients from cognitively unimpaired subjects and reflect cognitive measures and progression.142 P-tau181 levels have been studied in PD, and their ability to predict disease severity and cognitive decline has been mixed and, therefore, they cannot currently be recommended for trial use.143–145 Other tau species also show promise in AD and need further exploration in PD cohorts.

Neuroinflammation

Immune cells in the CNS and in the periphery are involved in PD neurodegeneration.146 Measurement of cellular components and levels of inflammatory mediators have, therefore, been explored for biomarker purposes (Table 2 ). Glial fibrillary acidic protein (GFAP) is released from astrocytes into the bloodstream, and its level can be used to distinguish PD from healthy controls,147,148 while its ability to discriminate PD from other atypical parkinsonisms is unclear. The glial activation biomarkers YKL-40 (chitinase-3-like protein 1) and MCP-1 (monocyte chemoattractant protein-1) are increased even further in atypical parkinsonian patients compared with PD and can thus reliably discriminate tauopathies from synucleinopathies,149,150 although this is best achieved by combining them with a panel of non-inflammatory CSF biomarkers [area under the curve (AUC) = 0.95].151 Among PD patients, GFAP levels seem to predict the development of dementia.152

Table 2.

Fluid and tissue biomarkers from aberrant pathways noted in Parkinson’s disease and their potential relevance to clinical trial design

Biomarker Origin Differentiating PD from healthy controls Marker of disease severity Differentiating PD from atypical parkinsonian disorders Predicting disease progression Surrogate for disease progression
Neuroinflammation
 Glial activation markers (YKL-40) CSF +++ +
 Glial activation markers (MCP-1) CSF +++ ++ +
 GFAP Serum/plasma + ++ +
 T-cell subtype level/ratios Blood +++ +++ +
 Neutrophil lymphocyte ratio Blood +++ +
 CRP Blood +++ + +
 Interleukin levels Blood +++ +++ ++
 TNF Blood +++ +++ +
 Complement levels Blood + +
 Chemokine ligand 5/RANTES Blood ++ ++
Lysosomal dysfunction
 Glucocerebrosidase activity CSF ++ + +
Blood ++
 β-hexosaminidase CSF +
 Cathepsin D CSF +
 Glucosylceramide CSF
Plasma ++
Serum
Mitochondrial dysfunction
 DJ-1 CSF + +
Plasma/serum + +
 Peroxisome proliferator-activated receptor γ coactivator 1α Blood ++ +
 Fibroblast growth factor 21 Serum
 Growth differentiation factor 15 Serum
Synaptic markers
 SNARE complex Plasma/serum (exosome) +
 SNAP25 CSF +
 Neurogranin CSF +++ ++
 β-Synuclein CSF +
 GAP43 CSF +
 Contactin-1 CSF + +
 Pentraxins CSF + + +
 Neurotransmitter levels CSF + +
 Dopamine metabolites (HVA, DOPAC) CSF +++ +++ +
Plasma ++ +
Axonal damage (NfL) CSF + +++ ++
Plasma/serum +++ +++ +++
Plasma/serum (exosome) +

DOPAC = dopamine, 3,4-dihydroxyphenylacetic acid; HVA = homovanillic acid; NfL = neurofilament light chain; PD = Parkinson’s disease. Grading approach adapted from Majbour et al.20

− = No effect (Also scored if negative in a meta-analysis).

+ = Effect 1 study/inconsistent results across studies.

++ = Effect in 2–3 studies using single site cohort.

+++ = Effect in ≥3 studies or multisite cohort (also scored if positive in meta-analysis).

Neutrophil-to-lymphocyte ratios (NLRs) are indicative of overall inflammatory status and are elevated in PD compared with healthy controls153 as is the proinflammatory lymphocyte profile (diminished T-regulatory and increased T-helper cell levels).154–157 The NLR has been negatively associated with presynaptic radionuclide striatal-binding ratios and positively associated with motor impairment,153,158,159 while a proinflammatory lymphocyte profile shift is associated with more severe motor and cognitive impairment,160,161 and an increase in Tregs expressing CD49d is linked to lesser motor impairment.162 Altered lymphocytes lead to and are in turn influenced by cytokines. Elevated C-reactive protein (CRP), interleukin (IL)-6 and IL-10 as well as tumour necrosis factor α and chemokine ligand 5 (CCL5, RANTES) levels have been noted in PD.163–171 Current evidence does not, however, suggest these markers would help in distinguishing PD from atypical conditions considering inconsistent findings between studies156,172–174 and small-to-intermediate effect sizes.175 Similarly, associations with non-motor symptoms noted particularly for IL-6 and IL-10176 are unlikely to be of value for trial design, although associations of pro-inflammatory cytokines, particularly CRP and CCL5, with reduced survival177 and the development of motor and cognitive impairment178–180 are of value for both prognosis and monitoring progression.

Taken together, the value of individual inflammatory markers is low, although combining several inflammatory markers to predict disease progression will likely contribute to future approaches.180,181 While better validated general biomarkers of progression exist, these panels could be particularly useful for enriching trials that test agents targeting inflammatory pathways.

Genetics and gene regulation

The relationship between genetic risk factors for PD and the pathophysiological processes underlying PD are under renewed scrutiny based on the use of α-syn SAA in CSF. People with leucine-rich repeat kinase 2 (LRRK2) mutations may develop typical PD, positive α-syn SAA in CSF and typical PD pathology at post-mortem,182 while the phenotype, pathophysiology and α-syn SAA findings and post-mortem pathology can also be completely different, despite the same LRRK2 mutation.183 The far lower rates of positivity of the CSF α-syn SAA among LRRK2 mutation carriers questions whether these patients should be included within trials specifically targeting α-syn and potentially other broad interventions being considered for PD neurodegeneration.184 Nevertheless, there is great interest in targeting LRRK2 as a means of influencing disease progression in PD, and genetic status may be of greater relevance for these interventions than other biomarkers. That said, the most advanced LRRK2 inhibitor trial has pragmatically chosen to focus recruitment of a combination of PD patients with and without LRRK2 mutations (NCT05348785), while other LRRK2-specific interventions may specifically want to recruit the subgroup who are positive for the α-syn SAA.

Of relevance to this point, molecular dysfunction of pathways downstream from LRRK2 also occur, and these are being explored as biomarkers in trials targeting this enzyme. pS1292-LRRK2 levels are higher in urinary EVs in idiopathic PD and correlate with motor severity.185 Furthermore, CSF EV pS1292-LRRK2 levels are 10-fold higher than urinary EV levels, suggesting relevance for CNS activity.186 Genetic variability may, therefore, be considered when selecting patients for precision medicine interventions as well as for helping to balance trial arms for progression or adjusting for baseline differences in longitudinal analysis. pS1292-LRRK2 levels or other downstream molecular abnormalities [whole-blood pS935 LRRK2 levels, peripheral blood mononuclear cell pT73 Rab10 levels, urine di-22:6-bis(monoacylglycerol) phosphate and CSF total LRRK2] may become useful tools for measuring target engagement and the therapeutic response to agents specifically targeting these pathways, as has been demonstrated in a recent early stage LRRK2 inhibitor trial187 (Supplementary Table 1).

Other genetic factors can also determine phenotypic severity and progression. PD patients with the A53T α-syn mutation experience worse autonomic and cognitive deterioration,188 while apolipoprotein E (APOE4) and glucosidase beta acid 1 (GBA1) PD patients have accelerated cognitive189–193 and motor deterioration,194 although this may be constrained to specific mutations/polymorphisms.195–197 Polygenic risk scores for predicting the rate of progression appear promising but need replication.198,199

Non-coding RNAs (ncRNA) contribute to gene expression regulation. Micro RNAs (miRNAs) are small ncRNAs (sncRNAs), which have been explored for biomarker potential. Unique serum miRNA patterns comprising upregulation (miR-6836-3p and miR-6777-3p) and downregulation (miR-493-5p, miR-487b-3p and miR-15b-5p) have been noted in PD200,201 and supported by known involvement of these miRNAs in PD pathogenic processes. Sampling, quantification and analysis approaches need to become standardized to facilitate between study comparisons. SncRNA analysis from CSF EVs may also be worth further exploration.202 While plasma EV miRNA measurement appears useful when distinguishing PD from healthy controls [AUC 0.85 (miR331-5p) and 0.90 (miR-505)203], the combination of miR153 and miR-409-3p using the CSF EV approach is even more impressive (AUC 0.99).204 miRNAs may likely play a diagnostic role in future trials depending on the mode of action of the drug being studied.

Lysosomal dysfunction

The GBA1 gene encodes the lysosomal enzyme β-glucocerebrosidase (GCase). GBA1 mutation carriers have almost uniformly positive α-syn SAA in CSF.4 Impaired GCase and other lysosomal enzyme activity [e.g. cathepsin D (CTSD)] in GBA1-carrier and non-carrier PD patients leads to lysosomal dysfunction, thus negatively impacting α-syn degradation.205,206 Although CSF GCase activity depends on the specific GBA1 mutation carried, levels are also lower in idiopathic PD patients compared with controls.207 GCase levels are, however, of low value for diagnosing PD, although combining GCase activity with oligomeric/total α-syn ratios (AUC = 0.87, 82% sensitivity, 71% specificity), as well as other lysosomal enzymes (CTSD and β-hexoxaminidase) and Aβ42, improves this (AUC = 0.83, 75% specificity, 84% sensitivity).208

CSF GCase levels correlate with cognitive impairment,209 while activity also seems to predict subsequent development of dementia regardless of genetic status.210 CSF GCase levels may, therefore, usefully allow enrichment of clinical trial arms testing agents targeting this enzyme (even in the absence of a GBA1 mutation) as well as a method for confirming target engagement. Blood GCase activity is also reduced compared with healthy controls, although prediction of progression has not been explored.211,212 GCase activity has been used as an exploratory outcome in recent disease modification trials in conjunction with its downstream hydrolytic product glucosylceramide (Supplementary Table 1). Glucosylceramide can distinguish GBA-PD from idiopathic PD and healthy controls and be measured in both plasma and peripheral blood mononuclear cells and therefore used as a biomarker for target engagement in clinical trials targeting GBA-PD.213,214

Mitochondrial dysfunction

Mitochondrial dysfunction contributes to the pathogenesis of PD.215 The existence of inherited autosomal recessive parkinsonism due to mutations of parkin (PRKN), PTEN induced kinase 1 (PINK1) and the protein deglycase (DJ-1) gene, which encode proteins that mediate mitophagy, supports this link.216,217 Typical α-syn pathology is less consistently reported in people with these mutations, and the rate of positivity of the α-syn SAA in CSF is also low,97,184 thus reinforcing the potential importance of both genetic testing and selection of additional other biomarkers during trial recruitment and follow-up, depending on the mode of action of the agent being tested.

The best explored mitochondrial biomarker in this context is CSF DJ-1, levels of which are decreased in PD218,219 compared with controls and correlate with disease severity,21 although similarities with other parkinsonian syndromes make its diagnostic use unlikely.220,221 Similar poor diagnostic value has been noted for serum and plasma DJ-1 levels.222–224 Other less well studied biomarkers include phosphorylated ubiquitin at the serine 65 residue (pSer65Ub), which occurs by virtue of the loss of mitochondrial membrane potential triggering the stabilization of PINK1 at the outer mitochondrial membrane.225 While increased pSer65Ub levels have been observed in PD post-mortem brains, lower levels have been identified in familial PD with PINK1/parkin mutations.226,227 Investigations of this marker in biofluid samples will be of interest, possibly as confirmation of target engagement and longitudinally to assess progression rates of disease in these PD subtypes. Similarly, the peroxisome proliferator-activated receptor γ coactivator 1 alpha (PGC-1α) has been of interest due to its role as a regulator of mitochondrial function.228 The PGC-1α reference gene and PGC-1α levels are downregulated in human brain and blood leucocytes in PD compared with control patients, and this negatively correlates with disease severity.229–231 Interventions targeting mitochondrial processes might usefully measure peripheral levels of PGC-1α.

A concern, however, for the use of mitochondrial blood-based biomarkers is that they do not recapitulate ‘neuronal’ mitochondrial dysfunction. Genetic mutations leading to mitochondrial dysfunction in PD often show tissue-specific expression patterns, and therefore peripheral blood changes may lack interpretability.232,233 This is supported by a recent study showing negligible diagnostic value for well-established biomarkers of mitochondrial disease such as fibroblast growth factor 21 and growth differentiation factor 15 in reflecting mitochondrial dysfunction in PD patients.226

Insulin resistance

The coexistence of type 2 diabetes mellitus (T2DM) with PD results in more rapid motor and cognitive progression.234–237 Faster progression appears to be independent from the existence of vascular disease in the brain238 and at least in part explained by disruptions in physiological brain insulin signalling (central insulin resistance)239 contributing to neurodegeneration.240

Central insulin resistance can be measured through abnormalities in insulin signalling mediated by insulin-receptor substrate-1 (IRS-1). Tyrosine IRS-1 phosphorylation (IRS-1 p-Tyr) evokes insulin responsiveness, while serine phosphorylation primarily deactivates IRS-1 and attenuates insulin signalling.239,241 Elevated IRS-1 phosphorylation at serine positions 616 (IRS-1 p-S616) and 312 (IRS-1 p-S312) represents attenuated insulin signalling242,243 and has been noted in plasma EVs of PD patients.244,245 Decreased IRS-1 p-Tyr distinguishes PD patients from healthy controls and predicts cognitive impairment and motor severity.141 Increases in EV IRS-1 p-Tyr were associated with motor benefits from exenatide in a clinical trial, while increases in downstream p-Akt S473 predicted treatment response (Supplementary Table 1).244

Peripheral insulin resistance as defined by a Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) value ≥ 2.0 or glycated haemoglobin (HbA1c) concentration ≥5.7%, occurs in up to 60% of PD patients.246 The mechanistic importance of these finding in PD remains unclear, as the HOMA-IR is not associated with cognition or motor symptoms.247,248 Abnormal range HbA1c levels, however, predict motor and cognitive severity and progression in PD, while also being associated with the degree of axonal damage.249–252 Further exploration of insulin resistance and/or body mass index in the selection of patients for trials of agents that mechanistically target this pathway is clearly of potential importance, while measurement of central insulin resistance using exosome IRS-1 p-Tyr may turn out to be of utility in confirming target engagement for a growing number of agents currently being studied for disease modification.253

Synaptic degeneration

Disruptions to vesicle-mediated trafficking and secretory pathways with downstream effects on neurotransmitter levels and signalling as well as synaptic plasticity are key features of synucleinopathies.254 Proteins at different levels of this process have been explored for biomarker use (Table 2). Evidence to date suggests limited usefulness in PD, in part due to the confounding effect of dopaminergic therapies. Despite some studies suggesting alterations in serum and CSF levels of synaptic dopamine potentiators [β-synuclein and growth associated protein 43 (GAP43)]254–260 and markers of synaptic plasticity [neurogranin (Ng), contactin-1 (CNTN-1) and the zinc transporter ZnT3] in PD, inconsistencies between studies and poor correlation with motor severity and cognitive progression make future utility unlikely.259,261–268

CSF concentrations of the secretory granule proteins (VGF and secretogranin-2) and the dense core vesicle protein prodynorphin are potentially useful in distinguishing PD from DLB or predicting cognitive decline.269,270 Similarly, preliminary studies suggest CSF levels of the excitatory-inhibitory regulatory protein, neuronal pentraxin-2 (NPTX2)270 and the glutamate receptor GluA3262 suggest value in reflecting cognitive status and distinguishing PD from DLB271 and thus warrant further exploration in the assessment of cognitive progression.

Measuring panels of CSF protein levels reflecting neurotransmitter secretion, synaptic plasticity and autophagy will likely shape any future use of these markers.272 An example of this approach includes combining CSF and serum EV levels of the principal components of the soluble N-ethylmaleimide sensitive factor attachment protein (SNARE) complex [synaptosomal-associated protein 25 (SNAP-25), the syntaxins 1A and 1B, syntaxin-binding protein-1 and the vesicle-associated membrane proteins (VAMP-1, VAMP-2)] with oligomeric α-syn to improve diagnostic accuracy.111,263 Similarly, combining CSF Ng, NPTX2, total α-syn and age273 or CNTN-1, total α-syn, total tau, phosphorylated tau and Aβ1-42261) can also improve diagnostic distinction.

A similar approach would also be worthwhile when considering the use of neurotransmitter metabolites. Despite decreased CSF levels of the dopamine metabolite homovanillic acid (HVA) being consistently noted in PD,274–279 repeated measurements in the Deprenyl and Tocopherol Antioxidative Therapy of Parkinsonism (DATATOP) study did not suggest usefulness for monitoring progression. Simultaneous metabolite panel measurement of dopaminergic [e.g. 3,4-dihydroxyphenylalanine (DOPA), dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC)], noradrenergic (e.g. 3,4-dihydroxyphenylglycol, 4-hydroxy-3-methoxyphenylglycol) and serotonergic [e.g. 5-hydroxy-3-indoleacetic acid (5-HIAA)] metabolites in CSF,278 however, correlates better with motor severity and dopamine transporter single-photon emission computed tomography (DAT-SPECT) uptake,280,281 and utility of the panel as a progression marker needs to be further explored.

Axonal damage

Neuro-axonal damage represents the end event in the pathophysiology of PD. Axon cytoskeletons are comprised of neurofilaments, structural proteins which allow for growth, with large, myelinated axons having the highest content.282 Neurofilament subunits are released upon axonal injury irrespective of the cause.282 The neurofilament light chain (NfL) subunit is of diagnostic value in degenerative parkinsonian syndromes,283 while also correlating with nigrostriatal degeneration and greater reductions in presynaptic putaminal DAT ratios over time.284,285 This said, CSF NfL concentration does not seem to be increased in early PD,286 and significant increases are more indicative of atypical diagnoses than PD.283,286–288

Blood NfL strongly correlates with CSF NfL289–291 and reflects neurodegeneration in PD.291–294 Although NfL levels were not elevated in a meta-analysis considering all patients with PD290 and in one study exploring EV NFL levels,295 levels seem to be higher in more advanced PD289,291,293,296 and the more severe postural instability and gait disorders (PIGD)-subtype.297,298 Consistent inverse associations with cognitive scores have been reported,48,292–294,299–302 while NfL levels also predict more severe motor progression,285 cognitive decline298,303 and progression to milestones [walking-aid, nursing-home living, reaching final Hoehn and Yahr (H&Y) Stage 5 or death]. Blood NfL may, therefore, be useful for trial stratification, although its potential use as a surrogate end point might depend on the disease stage of recruited participants and trial duration.296,304

The highest yield when using NfL seems to lie in combining it with clinical and disease-specific fluid biomarkers. Examples of this include the ratio of NfL to Aβ42 in CSF, discriminating PD from PSP with good accuracy (AUC 0.93, sensitivity 89%, specificity 93%)305 as well as the use of a stepwise approach of firstly distinguishing synucleinopathies from non-synucleinopathies with skin α-syn SAA and then further distinguishing MSA from PD with NfL306 or by combining CSF NfL, CSF α-syn SAA and brainstem imaging.307 Similarly, PD progression is better predicted when combining markers with serum NfL, genetic status (ApoE4 and GBA) and validated prognostic clinical variables (age, verbal fluency, Unified Parkinson’s Disease Rating Scale axial scores) predicting unfavourable progression better than individual markers.296

Imaging biomarkers

A range of imaging modalities have been explored for their biomarker potential (Table 3). These include sonographic measurement of nigral signal, imaging approaches that measure brain structure, spectroscopy to explore brain biochemical changes, functional imaging to measure connectivity changes and radionuclide imaging to assess pre- and postsynaptic dopaminergic and non-dopaminergic integrity as well as metabolic functional changes (Box 2). Each approach has its strengths and weaknesses, and potential biomarker roles in trials will depend on the stage of disease being studied as well as practical considerations of availability and effect strengths alongside and in comparison with fluid biomarkers.

Table 3.

Outlines the range of imaging biomarkers and their potential relevance to clinical trial design

Imaging modality Differentiating PD from healthy controls Marker of disease severity Differentiating PD from atypical parkinsonian disorders Predicting disease progression Surrogate for disease progression
Transcranial sonography + +
T1-weighted structural MRI ++ +++ +++ ++ +++
Neuromelanin MRI + + + ++
Iron sensitive MRI +++ + ++ + +++
Diffusion MRI +++ ++ ++ ++ ++
MR spectroscopy +++ ++ ++
Functional MRI ++ + +
PET/SPECT
 Radionuclide
 α-Syn
 Dopaminergic +++ +++ +++
 Non-dopaminergic ++ ++ ++
 Synaptic density ++ +
Metabolic and network imaging
 Glucose metabolism +++ + ++ ++ +
 Neuroinflammation + +

PD = Parkinson’s disease; SPECT = single-photon emission computed tomography. Grading approach adapted from Majbour et al.20

− = No effect (also scored if negative in a meta-analysis).

+ = Effect 1 study/inconsistent results across studies.

++ = Effect in 2–3 studies using single site cohort.

+++ = Effect in ≥3 studies or multisite cohort (also scored if positive in meta-analysis).

Box 2. Biomarker imaging techniques.

Transcranial sonography

  • ultrasound echogenicity measurement of brain tissues or structures through intact cranium—limited by lack of bone window in some subjects and inter-technician variability

Structural MRI

  • quantification of brain structural change using regions-of-interest or whole-brain approaches

  • commonly used sequences include T1, T2, T2*, R2* (R2* = 1/T2*)-weighted, susceptibility-weighted, proton-density-weighted, fluid-attenuated inversion recovery and neuromelanin-sensitive approaches

Proton magnetic resonance spectroscopy

  • estimates relative concentrations of proton-containing metabolites in brain

  • metabolites commonly assessed include N-acetylaspartate, choline-containing compounds, myo-inositol and creatine

Functional MRI

  • evaluates neuronal activity by measuring transient variations in blood flow and variation correlation in functionally connected regions

  • utilized under task-based or under resting-state conditions

Radiotracer imaging

  • measures pre- and postsynaptic receptor and transporter density as well as glucose metabolism and microglial activation using different radiotracers

  • provides information on nigrostriatal dopaminergic, serotonergic and cholinergic system integrity, regional tissue glucose metabolism and activity and status of microglial-mediated inflammation

In the proposed staging system for PD, the development of dopaminergic dysfunction has been incorporated as an important staging threshold.12 The range of imaging approaches that could be used for this are variable in their ability to discriminate PD from other pathophysiological processes as well as their potential for measuring the rate of progression of PD.

Transcranial sonographic imaging

Increased substantia nigra (SN) echogenicity, likely due to accumulation of nigral iron, is observed in PD,308–310 although a proportion of healthy controls and essential tremor patients also exhibit this.311 This sign can, however, differentiate PD from PSP and MSA with good sensitivity (91%) and specificity (82–96%).308 Hyper-echogenicity remains unchanged over follow-up312 and does not correlate with disease severity310,313 or presynaptic DAT loss,314 thus limiting use as a progression marker.

Structural MRI techniques

Structural MRI approaches comprise; T1-weighted structural imaging methods, which measure cortical and subcortical volumetric changes and brain atrophy; neuromelanin-sensitive T1-weighted imaging, which is sensitive to measuring neuromelanin-iron complexes; iron-sensitive MRI which captures iron deposition and dopaminergic cell loss; and diffusion imaging using either single-tensor or two-compartment diffusion modelling (free-water), which reflects neurodegeneration and/or neuroinflammation.

T1-weighted structural MRI

T1-based structural MRI methods comprise cortical thickness measurement, voxel-based morphometry (VBM) and deformation-based morphometry (DBM). Differences in these approaches are summarized in Box 2.

Structural differences in the midbrain, putamen, brainstem and cerebellum can distinguish PD from atypical parkinsonian disorders.315 This distinction is, however, best made in later disease stages at a time when disease modification approaches may be hardest to achieve. Novel automated indexes may improve this though will need to be tested in independent cohorts.316

In the PPMI cohort, deformation-based morphometry detected a unique atrophy pattern, which predicted motor progression in early PD without dementia.317 A faster decline in prefrontal and cingulate cortices and the caudate and thalamus has also been seen in de novo PD compared with controls,318 while greater frontal atrophy after 18 months has also been noted in PD patients without cognitive impairment with a disease duration of only 2 years319 (though these findings were separately contradicted320).

Studies in individuals with moderate to late-stage PD without dementia have also varied. No VBM differences were noted in one study,321 while another found reduced grey matter in the frontal lobe.322 Longitudinal atrophy of occipital and fusiform regions has been noted in patients with a disease duration of over 5 years without cognitive impairment, while patients with cognitive impairment develop greater and more widespread atrophy in supplementary motor area, temporal, parietal and occipital cortices.323 Accelerated loss of gyrification in bilateral frontal and parietal regions in patients with a disease duration greater than 5 years compared to less than 5 years has also been noted.324

In summary, T1-weighted structural MRI methods are sensitive to neurodegenerative progression even in the absence of cognitive impairment, although this also seems to be better in more advanced disease stages. Replication studies demonstrating patterns of atrophy progression depending on disease stages are however currently lacking and will be important before recommendation for trial use. Furthermore, ascertaining the precise role of ultra-high-field scanners (7 T and above), which can provide sub millimetric anatomical information and higher degrees of diagnostic detail compared with 3 T MRI,325 will be important. Planned future longitudinal studies will be critical for informing this.326

Neuromelanin and iron sensitive imaging

Neuromelanin imaging (NMI) demonstrates only moderate sensitivity and specificity for distinguishing PD from healthy controls,327–331 while signal differences are also suboptimal for distinguishing atypical parkinsonian conditions from PD.332,333 In contrast, however, NMI shows reduced signal across disease stages (disease duration of 1.5–10 years) with a ventrolateral to anteromedial SN progression pattern consistent with the neuropathological patterns of cell loss.

Iron-sensitive techniques including R2* relaxation imaging, susceptibility-weighted imaging (SWI) and quantitative susceptibility mapping (QSM) have similar ability to quantify nigral iron deposition as NMI.334–336 The absence of dorsal nigral hyperintensity corresponding to the region of nigrosome-1 (DNH) on iron-sensitive sequences distinguishes PD from controls well325,337,338 regardless of disease duration.339 Use for distinguishing atypical disorders from PD is however lacking, while progression marker use seems to be disease duration-dependent.

Although striatal, nigral, globus pallidus and caudate R2* relaxation rate increased in two separate studies after 2 years in early-stage PD,335,340 separate studies exploring R2* or QSM in de novo patients336 and patients with a disease duration <1 year showed no longitudinal changes.339 The use of R2* as a progression marker becomes clearer, however, in later disease stages,339 with increased relaxation time in SN R2* mapping over 3 years correlating with motor severity in cases with an initial disease duration of 5 years,341 while faster progression in the SN pars compacta seems to occur after a disease duration >5 years.339

Taken together, NMI and iron-sensitive imaging could potentially be developed usefully as progression biomarkers, although values will need to be considered in the context of disease duration. Obviously, the use of iron-sensitive modalities will be particularly advantageous in trials targeting iron.

Diffusion imaging

Although some studies have demonstrated reduced SN fractional anisotropy with single tensor diffusion imaging in early PD,342–344 this was not confirmed by a meta-analysis of 10 studies.345 Evidence in later disease (disease duration 10 years) is limited to one study demonstrating more anterior and rostral SN involvement.344 On balance, this approach cannot currently be recommended for progression marker use. The finding of diffusion abnormality of the nucleus basalis of Meynert predicting development of cognitive impairment could be explored for balancing arms in small trials or selecting phenotypes that are likely to respond to specific treatments though replication of this finding is important.346

Free water imaging studies have been more consistent with increased signal in the posterior SN being noted in early PD.347,348 Free water in the posterior SN also increases over 4 years and changes over 1 year can predict the H&Y 4-year change.348 This increase continues in later disease stages (duration over 7 years), where longitudinal increases in free water occur in the anterior but not posterior SN.349 This modality is promising as a progression biomarker but may require selection of the region of interest depending on the disease stage. Free water imaging of the basal ganglia, midbrain and cerebellum and the application of automated imaging differentiation is promising for differentiating PD from atypical conditions.350 This approach was found to be superior to a conventional magnetic resonance Parkinsonism index as well as plasma NfL levels for distinguishing PD from atypical conditions.351

Proton magnetic resonance spectroscopy

Proton magnetic resonance spectroscopy (MRS) reveals the metabolic status of the region sampled for a specific disease process. In PD, N-acetyl aspartate/creatine (NAA/Cr) ratios in the SN are reduced compared to controls and correlate with disease severity.352,353 Lower ratios have also been noted in the lentiform nucleus (LN), temporoparietal and posterior cingulate cortices, as well as the presupplementary motor area,354–357 although correlation with disease severity is less clear.355,356 NAA/Cr ratios are lower in the rostral SN in PD with an inverted pattern in atypical parkinsonian patients and healthy controls.358 Taken together, there is some preliminary level of evidence that MRS could serve to improve PD diagnostics, but it may be best used in combination with conventional MRI to increase specificity.

Phosphorus based MRS (31P-MRS) has been of specific interest for a subset of potential interventions as it can assess mitochondrial function. In vivo Pi/ATP and PCr/ATP ratios reflect oxidative phosphorylation pathways.359 Reductions in ATP and PCr360 and increased Pi/ATP ratios361 in the putamen and midbrain of PD patients compared with controls have been reported while differences can also distinguish PD from PSP (AUC 0.93).362 Longitudinal ratio improvement suggestive of target engagement was also noted in a recently completed disease modifying trial of ursodeoxycholic acid.363

Functional MRI

Resting-state and task-based functional MRI reveal networks involved in motor, cognitive and affective processes. Network impairments have been associated with motor and non-motor symptoms. Reduced resting-state connectivity between the striatum and thalamus, midbrain, pons and cerebellum has been noted in PD as have connectivity changes between cortical and subcortical areas.364 Reduced resting-state functional connectivity within the basal ganglia network can differentiate PD from healthy controls (sensitivity 100%, specificity 89.5%),365 while cerebellar connectivity with multiple brain networks differs between PD and MSA.366 Longitudinal task-based functional MRI can track progression with declining activity in the putamen and primary motor cortex over 1 year,367 although the impact of levodopa administration on network connectivity is an important consideration.368 While the available evidence for this modality is promising overall, more widespread replication of diagnostic and progression findings are necessary.

PET/SPECT imaging

Radionuclide imaging

Several radiolabelled probes for imaging α-syn have been explored though no tracer is currently of diagnostic value for PD. Issues to overcome include developing tracers for intracellular targeting with ideal lipophilicity and tracer selectivity for α-syn over amyloid and tau aggregates.369,370 More recently, however, a newly developed α-syn PET tracer, 18F-ACI-12589, was shown to bind to basal ganglia and cerebellar white matter in a small cohort, although this was confined to MSA patients.371 Larger studies examining diagnostic accuracy for distinguishing PD from MSA will be critical.

Dopaminergic tracers

A variety of radionuclide tracers are available to examine pre- and post-synaptic striatal dopaminergic function using PET or SPECT imaging. At the presynaptic level, molecular targets and their respective tracers include L-aromatic amino acid decarboxylase [AADC/tracer fluorodopa (F-DOPA)], vesicular monoamine transporter 2 (VMAT2/tracer 11C-dihydrotetrabenazine) and the DAT (DAT/tracers CFT PET and 123I-CIT SPECT) density.

These markers are sensitive for the detection of dysfunction or loss of striatal dopaminergic terminals and enable the identification of parkinsonian syndromes with nigral neurodegeneration but do not reliably distinguish PD from atypical disorders. Visual assessment for the presence of nigrostriatal degeneration with this modality is increasingly used in trial recruitment372 to exclude patients with clinical presentations in keeping with PD but with scans without evidence of dopaminergic deficit (SWEDDS) due to e.g. drug induced parkinsonism.373–375 Objective measurement of striatal uptake in comparison to other regions may, however, be more useful in trials recruiting patients with more established PD as these ratios can reflect motor and non-motor disease severity as well as progression through disease stages, although hemispheric dominance and type of tracer used are important considerations.376 Striatal dopaminergic markers decline most prominently in the first years of motor disease before largely plateauing within 5 years of diagnosis.377–380 Quantification of dopaminergic markers in the midbrain/SN may be better markers beyond this point.381

The type of dopaminergic tracer used can potentially be critical for tracking progression in trials and measuring treatment response with VMAT2 imaging is less subject to compensatory changes in expression than DAT and F-DOPA.382 Quantitative dopaminergic assessments have been used in a number of recent disease modification trials though with overall negative findings to date (Supplementary Table 1).

Dopamine receptor expression can also be estimated at the post-synaptic level with PET ligands such as 11C-raclopride, 18F-fallypride or 123I-IBZM SPECT (all of which bind to D2 receptors) or agents such as 11C-NNC 112, which binds to D(1) receptors.383 Preservation of post-synaptic dopamine receptors is typical of PD whereas post-synaptic receptor loss early in the disease is more likely indicative of an atypical form of parkinsonism. Imaging results depend on the dose and timing of oral dopaminergic replacement and the usefulness of this type of imaging approach may perhaps be restricted to restorative approaches such as cell or gene therapy interventions.384

Non-dopaminergic tracers

Radionuclide imaging studies of the serotonergic and cholinergic systems demonstrate associations with non-motor PD pathophysiology. Reduced binding on serotonergic imaging has been noted in individuals with early PD (disease duration less than 5 years).385 Serotonergic denervation also correlates with increased dopamine turnover and reduced levodopa responses.386 In later disease stages (disease duration 5–10 or more years), serotonergic transporter binding remains reduced compared to controls,385 and the degree of serotonergic pathology is associated with cognitive decline.387 Cholinergic denervation also occurs in early PD (disease duration less than 3 years) but is more pronounced in PD with dementia.388 Noradrenergic activity, quantifiable by PET imaging is reduced in PD and is associated with the presence of RBD and cognitive impairment.389 The utility of these markers in tracking progression is of interest but not yet sufficiently clear.

Synaptic density

Synaptic density quantification irrespective of neurotransmitter type has also been of interest in PD. Tracers quantifying the concentration of the synaptic vesicle 2A protein (18F-UCB-H or 11C-UCB-J) reflect this and have been studied in several cohorts. Lower binding potential in both cortical and subcortical regions have been noted in PD though this is most prominent in the SN.390 Correlation with clinical status has, however, been inconsistent though one study suggested more prominent and extensive reductions in PD dementia and DLB cases.391–393 Similarly, small cohort studies using 11C-UCB-J PET did not note binding changes over 2 years.391,394 Current evidence therefore does not support the use of this marker in clinical trials.

Metabolic and network imaging

Glucose metabolism

18F-FDG-PET parieto-occipital hypometabolism is noted in PD,395,396 while preserved glucose metabolism in the basal ganglia distinguishes PD from MSA and PSP.395 Inferior parietal and left caudate glucose hypometabolism in PD also correlates with motor and cognitive deficits.397 A unique PD-related pattern (PDRP) characterized by elevated pallidothalamic and pontine metabolic activity with reduction in the supplementary motor area, premotor cortex and parietal association areas has also been noted in cases prior to dopaminergic treatment398 and can differentiate PD from atypical parkinsonism.399

PDRP progresses in early PD (disease duration less than 2 years) over 24 months, suggesting potential progression marker use in the early stages,400 although a critical limitation is that acute dopaminergic treatment diminishes the pattern.401 A PD-related cognitive pattern (PDCP) characterized by a reduction in the medial frontal and parietal association regions and metabolic increase in cerebellar cortex and dentate nuclei402 has also been described. This pattern seems to occur years after the PDRP,400,403 increases over time400 and is higher in those with dementia.404 The PDCP also correlates with memory and executive performance,402 while its lack of change with dopaminergic treatment potentially supports its use as a marker of cognitive dysfunction.405 These separate metabolic networks could potentially be used to track progression and treatment response in the appropriate setting.

Neuroinflammation imaging

The PET ligands 11C-PK11195, 11C-PBR28 and 18F-FEPPA, which bind to the 18 kDa translocator protein (TSPO) on mitochondria in microglia, have been used for imaging neuroinflammation with TSPO upregulation suggesting microglial activation.406

PD clinical severity and putaminal presynaptic dopaminergic integrity correlates with 11C-PK11195 binding.407 Binding affinity can vary with TPSO genetic polymorphisms which needs appropriate adjustment in analyses.406,408 Taken alone, TPSO patterns lack the ability to distinguish parkinsonian conditions though their future use may be as biomarkers of therapeutic response for interventions targeting neuroinflammation.409

Limitations of biomarkers

A framework for considering the definition of PD according to the presence/absence of α-syn SAA-CSF is potentially a major step forward in planning PD trials. Several practical obstacles need to be considered however prior to the routine use/reliance on biomarkers in the clinical trial context. Firstly, acquiring some biomarkers, e.g. CSF, requires an invasive procedure, which may be unacceptable for some participants. Growing evidence of the equivalence of α-syn SAA-in skin to that seen with CSF could, however, overcome this limitation. The demonstration of equivalence of testing on even less invasive samples such as serum/plasma or within peripherally obtained EVs is therefore a priority. With greater demonstration of validity, routine testing of peripherally acquired biomarkers can become normal practice, for example the widespread availability of plasma NfL testing in healthcare laboratories.

Interpretation of discrepant results between studies attributable to preanalytical and analytical confounders, different techniques employed and a lack of factoring of different protein species measured (total α-syn versus oligomeric) needs careful critique. Similarly, imaging studies are affected by methodological discrepancies, including different assumptions for correction of serial data as well as sample size, power and study design caveats and the use of different outcome measures. Collaborative studies allowing analysis of larger sample sizes with adequate follow-up that employ standardized sampling and analysis methodology will improve these limitations, as demonstrated by the harmonization of large numbers of samples processed in the PPMI.

The major limitation in biomarker discovery is undoubtedly difficulty with validation. Association between a change in a biological assay alongside a clinical state need not equal causation. For example, biological changes may represent healthy compensatory responses to a pathological process. Furthermore, even biomarkers that do reflect active processes of neurodegeneration may not have linear relationships over the course of disease particularly if production ultimately declines because of widespread tissue death. While it is possible to use clinico-pathological data for validation, confirmation that a biomarker predicts slowing of disease progression necessarily requires the identification of an agent which achieves this according to our threshold, whether that be clinical, patient reported, functional impairment or quality of life milestones, which have inherent limitations.

To date, no single biomarker can yet be recommended to act as a surrogate for clinical disease progression in PD. Combinations of fluid biomarkers invariably increase the strength of their individual predictive properties. While fluid and imaging biomarkers are often collected from the same trial participants, explorations of the utility of multiple fluid biomarkers as a panel alongside imaging in combination are rare. This approach was partly adopted in the recent deferiprone trial (Supplementary Table 1) where brain iron content using T2* sequences and plasma ferritin and prolactin levels were used as combined markers of target engagement and specific measures of treatment effect while structural imaging for measurement of brain atrophy and DAT-SPECT imaging was used to explore the impact of the agent on overall disease progression (atrophy and nigrostriatal degeneration). Although clinical worsening in the deferiprone treated group complicates interpretation of how well the panel of biomarkers performed, one could argue that they did reflect the effect of the drug with decreased nigrostriatal iron content and plasma ferritin and increased plasma prolactin in the deferiprone group, while no inverse correlation between brain-structure volumes and iron content was noted in keeping with the negative clinical findings over a relatively short duration of follow-up.

Challenges for future trials will be in the choice of selection of suitable combinations of fluid and imaging biomarkers that complement each other. This will certainly need to be strongly guided by the biological action of the agent being tested and the stage of the disease of their participants being treated, although those biomarkers that appear to align most closely with disease progression should be prioritized. How much weight each biomarker in the panel will ultimately carry will become more easily evident following a positive clinical trial.

Conclusions and recommendations

The identification of a better framework for the certainty of a PD diagnosis based on positivity of α-syn SAA-CSF is a major step forward, and less invasive equivalent alternatives will help even more. The further development of reliable biomarkers of PD neurodegeneration could further facilitate prognostication, identification of disease subtypes, conduct of clinical trials and identification of agents that may slow down or stop these processes. The precise role for biomarkers will depend on the mechanism of action of the agent in question, and the decision made regarding the stage of the illness at which the intervention is being applied. There is interest in recruiting people earlier in the neurodegenerative process, even prior to symptom onset, given that, intuitively, earlier intervention may provide a better chance of preventing irreversible cell death.410 Alongside trials in prodromal cohorts, there will remain a need to identify whether any disease modifying intervention has an impact on the 6–10 million people already struggling with symptoms and in need of prevention of further decline.

In this group, PD diagnosis is less difficult though a sizeable proportion of cases at this stage with atypical parkinsonian disorders can be mistaken as suffering from PD and therefore inadvertently recruited into disease modifying trials. While there will remain healthy debate whether α-syn oligomeric seeding and propagation is the primary cause of PD neurodegeneration, it appears that the α-syn SAA-CSF assay reflects an α-syn-related neurodegenerative process and can reliably distinguish synucleinopathies from other causes of parkinsonism/tremor with high specificity.

PD subtyping is also a high priority for better selection of responders. For example, interventions that specifically target an aspect of disease pathophysiology associated with genetic abnormalities could be specifically tailored to these patients.411 Mutations in GBA1 confer a worse prognosis and therefore a trial enriched with these patients may potentially allow an earlier signal of efficacy. In parallel, enhancement of GCase activity may also have therapeutic benefits in PD patients without GBA1 mutations.412

Features that strongly predict subsequent disease progression need to be carefully considered during treatment allocation. The randomization process itself should lead to balancing of features between placebo and active treatment arms, however this can fail to achieve this in smaller sized trials. The application of a panel of biomarkers for example pro-inflammatory immune markers which predict faster progression180 and reflect different aspects of disease-related pathways would be a useful approach to stratify patients into prognostic groups and potential responders to the treatment being tested, which will in turn enable more efficient and cost-effective collection of data and increase the likelihood of detecting an effect.

The most useful function of biomarkers is in the prediction that a change in any such biomarker reliably predicts slowing down of the neurodegenerative process that translates to reduction in disability accrual, and maintenance of function and quality of life. Towards this, the ratio of phosphorylated or oligomeric α-syn to total α-syn in CSF appears to be an encouraging fluid biomarker for disease progression. Technical challenges notwithstanding, measurement of one or both of these ratios may become routine practice in clinical trials of disease modifying agents to further improve diagnostic precision at baseline, minimize difference between trial arms and monitor changes in response to the intervention. The selection of a single fluid biomarker is likely to be a lower sensitivity surrogate for disease progression than the use of a panel of biomarkers. The development of a poly-biomarker, analogous to a polygenic risk score, will require careful modelling in large cohorts that have collected identical panels using agreed standardized operating procedures for their collection.

There are several structural imaging techniques that seem to track disease progression in PD reliably, perhaps the most useful of which are neuromelanin or free water MRI. Whether these allow sufficient resolution to quantify changes over shorter time periods than needed for conventional clinical methods, requires further data. Functional or PET imaging may allow more rapid confirmation of target engagement in trials, and their routine use may depend on the putative mechanism of action of the intervention, e.g. TSPO PET, in a trial of a neuroinflammatory intervention. While stabilization of fluid, imaging or tissue biomarkers should mirror attenuation of α-syn aggregation within the brain, it remains to be seen whether change in biomarker activity can reliably predict subsequent clinical disease progression.

In terms of recommendations, during the design and conduct of a clinical trial of a disease modifying intervention in PD, we suggest:

  • For broad interventions, investigators should routinely collect a biomarker (CSF, skin, blood) that can be used for an α-syn SAA as part of the trial inclusion criteria. Currently, SAA offers the highest specificity in distinguishing PD from controls or PD-like conditions, but it’s utility in differentiating PD from MSA requires further assay refinement.

  • For precision interventions, investigators should consider whether the planned intervention targets an alternative process that can be defined by an alternative genetic marker (LRRK2, GBA1, mitochondrial mutation) or measurable pathophysiological process (neuroinflammation, bioenergetics), irrespective of α-syn SAA.

  • Investigators should consider incorporating such a biomarker within the trial inclusion criteria, while also ensuring the biomarker is appropriate for the stage of disease being studied.

  • Where appropriate, the same biomarker might also be used to confirm target engagement of the intervention.

  • Clinical outcome analyses may need to incorporate baseline differences in panels of wet biomarkers as well as imaging differences between treatment groups predictive of more rapid progression.

  • Investigators should formally evaluate the relationship between biomarker changes and predicting the clinical effect of the intervention.

  • Consideration should be given at an early stage how biomarker data can be usefully shared/integrated to maximize learning across interventions.

Until we have identified an agent that slows down clinical progression, it will be difficult to conclude the validity of any biomarker at predicting such disease modification. It appears as a somewhat circular argument therefore, that we need success, before we can be confident in our tools designed to help achieve success. Faced with this challenge, the most practical path forward is to systematically collect specimens from participants in clinical trials for future research, while also incorporating longitudinal measurement of encouraging biomarkers for continued comparison with clinical progression measures. This requires a degree of consensus in the PD trials community regarding standardized protocols for specimen collection and analysis. The Critical Path for Parkinson’s (CPP) consortium is helping to achieve this.413 Differences in the longitudinal change in biomarkers according to candidate interventions will undoubtedly help in the understanding of target engagement and help in the eventual prediction of long-term outcomes and ultimately are likely to become reliable surrogate outcome measures.

In conclusion, we should remain optimistic that the use of a combination of fluid, tissue and imaging biomarkers may become sufficient to reliably demonstrate disease modification. There is already a precedent that change in an imaging biomarker has been considered sufficient evidence, by some, to conclude disease modifying properties of aducanumab in AD.414 This decision has been controversial, and it is likely that a more robust conclusion in PD would only be reached once any combination of biomarkers has been comprehensively validated in relation to patient reports of clinical symptoms of relevance to their health and wellbeing. In the meantime, the best biomarker candidates can already likely improve the selection of participants and may contribute to early assessments of target engagement and of efficacy in counteracting pathophysiological mechanisms. An ongoing systematic process of confirming clinico-biomarker validity and utility is required.

Supplementary Material

awad265_Supplementary_Data

Acknowledgements

T.F. has received grants from National Institute of Health Research, Edmond J Safra Foundation, Michael J Fox Foundation, John Black Charitable Foundation, Cure Parkinson’s, Innovate UK, Van Andel Research Institute and Defeat MSA.

Contributor Information

Nirosen Vijiaratnam, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.

Thomas Foltynie, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.

Funding

N.V.’s research time and position is funded by the Janet Owens Legacy fund. This research was supported by the National Institute for Health Research University College London Hospitals Biomedical Research Centre. The UCL Movement Disorders Centre is supported by the Edmond J. Safra Philanthropic Foundation.

Competing interests

The authors report no competing interests.

Supplementary material

Supplementary material is available at Brain online.

References

  • 1. Vijiaratnam N, Simuni T, Bandmann O, Morris HR, Foltynie T. Progress towards therapies for disease modification in Parkinson’s disease. Lancet Neurol. 2021;20:559–572. [DOI] [PubMed] [Google Scholar]
  • 2. Biomarkers Definitions Working Group . Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69:89–95. [DOI] [PubMed] [Google Scholar]
  • 3. Oueslati A, Fournier M, Lashuel HA. Role of post-translational modifications in modulating the structure, function and toxicity of alpha-synuclein: Implications for Parkinson’s disease pathogenesis and therapies. Prog Brain Res. 2010;183:115–145. [DOI] [PubMed] [Google Scholar]
  • 4. Siderowf A, Concha-Marambio L, Lafontant DE, et al. Assessment of heterogeneity among participants in the Parkinson’s progression markers initiative cohort using alpha-synuclein seed amplification: A cross-sectional study. Lancet Neurol. 2023;22:407–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Koga S, Sekiya H, Kondru N, Ross OA, Dickson DW. Neuropathology and molecular diagnosis of synucleinopathies. Mol Neurodegener. 2021;16:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Fenyi A, Leclair-Visonneau L, Clairembault T, et al. Detection of alpha-synuclein aggregates in gastrointestinal biopsies by protein misfolding cyclic amplification. Neurobiol Dis. 2019;129:38–43. [DOI] [PubMed] [Google Scholar]
  • 7. Kang UJ, Boehme AK, Fairfoul G, et al. Comparative study of cerebrospinal fluid alpha-synuclein seeding aggregation assays for diagnosis of Parkinson’s disease. Mov Disord. 2019;34:536–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Manne S, Kondru N, Jin H, et al. Blinded RT-QuIC analysis of alpha-synuclein biomarker in skin tissue from Parkinson’s disease patients. Mov Disord. 2020;35:2230–2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Rossi M, Candelise N, Baiardi S, et al. Ultrasensitive RT-QuIC assay with high sensitivity and specificity for Lewy body-associated synucleinopathies. Acta Neuropathol. 2020;140:49–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Russo MJ, Orru CD, Concha-Marambio L, et al. High diagnostic performance of independent alpha-synuclein seed amplification assays for detection of early Parkinson’s disease. Acta Neuropathol Commun. 2021;9:179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Bellomo G, De Luca CMG, Paoletti FP, Gaetani L, Moda F, Parnetti L. alpha-Synuclein seed amplification assays for diagnosing synucleinopathies: The way forward. Neurology. 2022;99:195–205. [DOI] [PubMed] [Google Scholar]
  • 12. Chahine LM, Merchant K, Siderowf A, et al. Proposal for a biologic staging system of Parkinson’s disease. J Parkinsons Dis. 2023;13:297–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Höglinger GU, Adler CH, Berg D, et al. Towards a biological definition of Parkinson’s disease. Preprints. [Preprint] 10.20944/preprints202304.0108.v1 [DOI] [Google Scholar]
  • 14. Reinoso G, Allen JC Jr, Au WL, Seah SH, Tay KY, Tan LC. Clinical evolution of Parkinson’s disease and prognostic factors affecting motor progression: 9-year follow-up study. Eur J Neurol. 2015;22:457–463. [DOI] [PubMed] [Google Scholar]
  • 15. Papapetropoulos S. Patient diaries as a clinical endpoint in Parkinson’s disease clinical trials. Cns Neurosci Ther. 2012;18:380–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Utsumi H, Terashi H, Ishimura Y, et al. How far do the complaints of patients with Parkinson’s disease reflect motor fluctuation? Quantitative analysis using a portable gait rhythmogram. ISRN Neurol. 2012;2012:372030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Davidson MB, McGhee DJ, Counsell CE. Comparison of patient rated treatment response with measured improvement in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2012;83:1001–1005. [DOI] [PubMed] [Google Scholar]
  • 18. Parashos SA, Luo S, Biglan KM, et al. Measuring disease progression in early Parkinson disease the National Institutes of Health Exploratory Trials in Parkinson disease (NET-PD) experience. JAMA Neurol. 2014;71:710–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. McGhee D, Parker A, Fielding S, Counsell C. Which clinical measures are most appropriate for measuring disease progression in Parkinson’s disease? J Neurol Neurosur Psychiatry. 2013;84:e2.72–e2. [Google Scholar]
  • 20. Majbour N, Aasly J, Abdi I, et al. Disease-associated alpha-synuclein aggregates as biomarkers of Parkinson disease clinical stage. Neurology. 2022;99:e2417–e2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Hong Z, Shi M, Chung KA, et al. DJ-1 and alpha-synuclein in human cerebrospinal fluid as biomarkers of Parkinson’s disease. Brain. 2010;133(3):713–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel-Doring F, Trenkwalder C. Schlossmacher MG. Alpha-synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: A cohort study. Lancet Neurol. 2011;10:230–240. [DOI] [PubMed] [Google Scholar]
  • 23. Mollenhauer B, Trautmann E, Taylor P, et al. Total CSF alpha-synuclein is lower in de novo Parkinson patients than in healthy subjects. Neurosci Lett. 2013;532:44–48. [DOI] [PubMed] [Google Scholar]
  • 24. Tokuda T, Salem SA, Allsop D, 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] [PubMed] [Google Scholar]
  • 25. Eusebi P, Giannandrea D, Biscetti L, et al. Diagnostic utility of cerebrospinal fluid alpha-synuclein in Parkinson’s disease: A systematic review and meta-analysis. Mov Disord. 2017;32:1389–1400. [DOI] [PubMed] [Google Scholar]
  • 26. Gao L, Tang H, Nie K, et al. Cerebrospinal fluid alpha-synuclein as a biomarker for Parkinson’s disease diagnosis: A systematic review and meta-analysis. Int J Neurosci. 2015;125:645–654. [DOI] [PubMed] [Google Scholar]
  • 27. Sako W, Murakami N, Izumi Y, Kaji R. Reduced alpha-synuclein in cerebrospinal fluid in synucleinopathies: Evidence from a meta-analysis. Mov Disord. 2014;29:1599–1605. [DOI] [PubMed] [Google Scholar]
  • 28. Zhou B, Wen M, Yu WF, Zhang CL, Jiao L. The diagnostic and differential diagnosis utility of cerebrospinal fluid alpha -synuclein levels in Parkinson’s disease: A meta-analysis. Parkinsons Dis. 2015;2015:567386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. De Bartolo MI, Vivacqua G, Belvisi D, et al. A combined panel of salivary biomarkers in de novo Parkinson’s disease. Ann Neurol. 2023;93:446–459. [DOI] [PubMed] [Google Scholar]
  • 30. Chahine LM, Beach TG, Brumm MC, et al. In vivo distribution of alpha-synuclein in multiple tissues and biofluids in Parkinson disease. Neurology. 2020;95:e1267–e1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Stuendl A, Kunadt M, Kruse N, et al. Induction of alpha-synuclein aggregate formation by CSF exosomes from patients with Parkinson’s disease and dementia with Lewy bodies. Brain. 2016;139(2):481–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Shi M, Liu C, Cook TJ, et al. Plasma exosomal alpha-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol. 2014;128:639–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Stuendl A, Kraus T, Chatterjee M, et al. alpha-Synuclein in plasma-derived extracellular vesicles is a potential biomarker of Parkinson’s disease. Mov Disord. 2021;36:2508–2518. [DOI] [PubMed] [Google Scholar]
  • 34. Zhao ZH, Chen ZT, Zhou RL, Zhang X, Ye QY, Wang YZ. Increased DJ-1 and alpha-synuclein in plasma neural-derived exosomes as potential markers for Parkinson’s disease. Front Aging Neurosci. 2019;10:438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Niu M, Li Y, Li G, et al. A longitudinal study on alpha-synuclein in plasma neuronal exosomes as a biomarker for Parkinson’s disease development and progression. Eur J Neurol. 2020;27:967–974. [DOI] [PubMed] [Google Scholar]
  • 36. Jiang C, Hopfner F, Katsikoudi A, et al. Serum neuronal exosomes predict and differentiate Parkinson’s disease from atypical parkinsonism. J Neurol Neurosurg Psychiatry. 2020;91:720–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Fu Y, Jiang C, Tofaris GK, Davis JJ. Facile impedimetric analysis of neuronal exosome markers in Parkinson’s disease diagnostics. Anal Chem. 2020;92:13647–13651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Dutta S, Hornung S, Kruayatidee A, et al. alpha-Synuclein in blood exosomes immunoprecipitated using neuronal and oligodendroglial markers distinguishes Parkinson’s disease from multiple system atrophy. Acta Neuropathol. 2021;142:495–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Si X, Tian J, Chen Y, Yan Y, Pu J, Zhang B. Central nervous system-derived exosomal alpha-synuclein in serum may be a biomarker in Parkinson’s disease. Neuroscience. 2019;413:308–316. [DOI] [PubMed] [Google Scholar]
  • 40. Cao Z, Wu Y, Liu G, et al. alpha-Synuclein in salivary extracellular vesicles as a potential biomarker of Parkinson’s disease. Neurosci Lett. 2019;696:114–120. [DOI] [PubMed] [Google Scholar]
  • 41. Zhao Y, Yang GF. Potential of extracellular vesicles in the Parkinson’s disease-pathological mediators and biomarkers. Neurochem Int. 2021;144:104974. [DOI] [PubMed] [Google Scholar]
  • 42. Ohmichi T, Mitsuhashi M, Tatebe H, Kasai T, El-Agnaf OMA, Tokuda T. Quantification of brain-derived extracellular vesicles in plasma as a biomarker to diagnose Parkinson’s and related diseases. Parkinsonism Relat Dis. 2019;61:82–87. [DOI] [PubMed] [Google Scholar]
  • 43. Xylaki M, Chopra A, Weber S, Bartl M, Outeiro TF, Mollenhauer B. Extracellular vesicles for the diagnosis of Parkinson’s disease: Systematic review and meta-analysis. Mov Disord. 2023;38:1585–1597. [DOI] [PubMed] [Google Scholar]
  • 44. Jiang C, Hopfner F, Berg D, et al. Validation of alpha-synuclein in L1CAM-immunocaptured exosomes as a biomarker for the stratification of Parkinsonian syndromes. Mov Disord. 2021;36:2663–2669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Wang Y, Shi M, Chung KA, et al. Phosphorylated alpha-synuclein in Parkinson’s disease. Sci Transl Med. 2012;4:121ra20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Stewart T, Sossi V, Aasly JO, et al. Phosphorylated alpha-synuclein in Parkinson’s disease: Correlation depends on disease severity. Acta Neuropathol Commun. 2015;3:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Majbour NK, Vaikath NN, van Dijk KD, et al. Oligomeric and phosphorylated alpha-synuclein as potential CSF biomarkers for Parkinson’s disease. Mol Neurodegener. 2016;11:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Oosterveld LP, Verberk IMW, Majbour NK, et al. CSF Or serum neurofilament light added to alpha-synuclein panel discriminates Parkinson’s from controls. Mov Disord. 2020;35:288–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Parnetti L, Gaetani L, Eusebi P, et al. CSF And blood biomarkers for Parkinson’s disease. Lancet Neurol. 2019;18:573–586. [DOI] [PubMed] [Google Scholar]
  • 50. Cariulo C, Martufi P, Verani M, et al. Phospho-S129 alpha-synuclein is present in human plasma but not in cerebrospinal fluid as determined by an ultrasensitive immunoassay. Front Neurosci. 2019;13:889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Foulds PG, Mitchell JD, Parker A, et al. Phosphorylated alpha-synuclein can be detected in blood plasma and is potentially a useful biomarker for Parkinson’s disease. FASEB J. 2011;25:4127–4137. [DOI] [PubMed] [Google Scholar]
  • 52. Foulds PG, Diggle P, Mitchell JD, et al. A longitudinal study on alpha-synuclein in blood plasma as a biomarker for Parkinson’s disease. Sci Rep. 2013;3:2540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Foulds PG, Yokota O, Thurston A, et al. Post mortem cerebrospinal fluid alpha-synuclein levels are raised in multiple system atrophy and distinguish this from the other alpha-synucleinopathies, Parkinson’s disease and dementia with Lewy bodies. Neurobiol Dis. 2012;45:188–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Majbour NK, Aasly JO, Hustad E, et al. CSF Total and oligomeric alpha-synuclein along with TNF-alpha as risk biomarkers for Parkinson’s disease: A study in LRRK2 mutation carriers. Transl Neurodegener. 2020;9:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. van Steenoven I, Majbour NK, Vaikath NN, et al. alpha-Synuclein species as potential cerebrospinal fluid biomarkers for dementia with Lewy bodies. Mov Disord. 2018;33:1724–1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Schulz I, Kruse N, Gera RG, et al. Systematic assessment of 10 biomarker candidates focusing on alpha-synuclein-related disorders. Mov Disord. 2021;36:2874–2887. [DOI] [PubMed] [Google Scholar]
  • 57. Ikemura M, Saito Y, Sengoku R, et al. Lewy Body pathology involves cutaneous nerves. J Neuropathol Exp Neurol. 2008;67:945–953. [DOI] [PubMed] [Google Scholar]
  • 58. Wang N, Gibbons CH, Lafo J, Freeman R. alpha-Synuclein in cutaneous autonomic nerves. Neurology. 2013;81:1604–1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Wang N, Garcia J, Freeman R, Gibbons CH. Phosphorylated alpha-synuclein within cutaneous autonomic nerves of patients with Parkinson’s disease: The implications of sample thickness on results. J Histochem Cytochem. 2020;68:669–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Liu X, Yang J, Yuan Y, et al. Optimization of the detection method for phosphorylated alpha-synuclein in Parkinson disease by skin biopsy. Front Neurol. 2020;11:569446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Donadio V, Incensi A, El-Agnaf O, et al. Skin alpha-synuclein deposits differ in clinical variants of synucleinopathy: An in vivo study. Sci Rep. 2018;8:14246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Gibbons C, Wang N, Rajan S, et al. Cutaneous alpha-synuclein signatures in patients with multiple system atrophy and Parkinson disease. Neurology. 2023;100:e1529–e1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Donadio V, Incensi A, Rizzo G, et al. Skin biopsy may help to distinguish multiple system atrophy-Parkinsonism from Parkinson’s disease with orthostatic hypotension. Movement Disord. 2020;35:1649–1657. [DOI] [PubMed] [Google Scholar]
  • 64. Kang W, Chen W, Yang Q, et al. Salivary total alpha-synuclein, oligomeric alpha-synuclein and SNCA variants in Parkinson’s disease patients. Sci Rep. 2016;6:28143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Tokuda T, Qureshi MM, Ardah MT, et al. Detection of elevated levels of alpha-synuclein oligomers in CSF from patients with Parkinson disease. Neurology. 2010;75:1766–1772. [DOI] [PubMed] [Google Scholar]
  • 66. Hansson O, Hall S, Ohrfelt A, et al. Levels of cerebrospinal fluid alpha-synuclein oligomers are increased in Parkinson’s disease with dementia and dementia with Lewy bodies compared to Alzheimer’s disease. Alzheimers Res Ther. 2014;6:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Park MJ, Cheon SM, Bae HR, Kim SH, Kim JW. Elevated levels of alpha-synuclein oligomer in the cerebrospinal fluid of drug-naive patients with Parkinson’s disease. J Clin Neurol. 2011;7:215–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Parnetti L, Farotti L, Eusebi P, et al. Differential role of CSF alpha-synuclein species, tau, and Abeta42 in Parkinson’s disease. Front Aging Neurosci. 2014;6:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. El-Agnaf OM, Salem SA, Paleologou KE, 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] [PubMed] [Google Scholar]
  • 70. Duran R, Barrero FJ, Morales B, Luna JD, Ramirez M, Vives F. Plasma alpha-synuclein in patients with Parkinson’s disease with and without treatment. Mov Disord. 2010;25:489–493. [DOI] [PubMed] [Google Scholar]
  • 71. Papagiannakis N, Koros C, Stamelou M, et al. Alpha-synuclein dimerization in erythrocytes of patients with genetic and non-genetic forms of Parkinson’s disease. Neurosci Lett. 2018;672:145–149. [DOI] [PubMed] [Google Scholar]
  • 72. Daniele S, Frosini D, Pietrobono D, et al. alpha-Synuclein heterocomplexes with beta-amyloid are increased in red blood cells of Parkinson’s disease patients and correlate with disease severity. Front Mol Neurosci. 2018;11:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Vivacqua G, Latorre A, Suppa A, et al. Abnormal salivary total and oligomeric alpha-synuclein in Parkinson’s disease. PLoS One. 2016;11:e0151156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Vivacqua G, Suppa A, Mancinelli R, et al. Salivary alpha-synuclein in the diagnosis of Parkinson’s disease and progressive supranuclear palsy. Parkinsonism Relat Disord. 2019;63:143–148. [DOI] [PubMed] [Google Scholar]
  • 75. Devic I, Hwang H, Edgar JS, et al. Salivary alpha-synuclein and DJ-1: Potential biomarkers for Parkinson’s disease. Brain. 2011;134(Pt 7):e178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Kharel S, Ojha R, Bist A, Joshi SP, Rauniyar R, Yadav JK. Salivary alpha-synuclein as a potential fluid biomarker in Parkinson’s disease: A systematic review and meta-analysis. Aging Med (Milton). 2022;5:53–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Hamm-Alvarez SF, Okamoto CT, Janga SR, et al. Oligomeric alpha-synuclein is increased in basal tears of Parkinson’s patients. Biomark Med. 2019;13:941–952. [DOI] [PubMed] [Google Scholar]
  • 78. Maass F, Rikker S, Dambeck V, et al. Increased alpha-synuclein tear fluid levels in patients with Parkinson’s disease. Sci Rep. 2020;10:8507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Yanamandra K, Gruden MA, Casaite V, Meskys R, Forsgren L, Morozova-Roche LA. alpha-Synuclein reactive antibodies as diagnostic biomarkers in blood sera of Parkinson’s disease patients. PLoS One. 2011;6:e18513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Wang X, Yu S, Li F, Feng T. Detection of alpha-synuclein oligomers in red blood cells as a potential biomarker of Parkinson’s disease. Neurosci Lett. 2015;599:115–119. [DOI] [PubMed] [Google Scholar]
  • 81. Meloni M, Agliardi C, Guerini FR, et al. Oligomeric alpha-synuclein and tau aggregates in NDEVs differentiate Parkinson’s disease from atypical parkinsonisms. Neurobiol Dis. 2023;176:105947. [DOI] [PubMed] [Google Scholar]
  • 82. Schweighauser M, Shi Y, Tarutani A, et al. Structures of alpha-synuclein filaments from multiple system atrophy. Nature. 2020;585:464–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Yang Y, Shi Y, Schweighauser M, et al. Structures of alpha-synuclein filaments from human brains with Lewy pathology. Nature. 2022;610:791–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Parnetti L, Chiasserini D, Persichetti E, et al. Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in Parkinson’s disease. Mov Disord. 2014;29:1019–1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Zheng H, Xie Z, Zhang X, et al. Investigation of alpha-synuclein species in plasma exosomes and the oligomeric and phosphorylated alpha-synuclein as potential peripheral biomarker of Parkinson’s disease. Neuroscience. 2021;469:79–90. [DOI] [PubMed] [Google Scholar]
  • 86. Fairfoul G, McGuire LI, Pal S, et al. Alpha-synuclein RT-QuIC in the CSF of patients with alpha-synucleinopathies. Ann Clin Transl Neurol. 2016;3:812–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Poggiolini I, Gupta V, Lawton M, et al. Diagnostic value of cerebrospinal fluid alpha-synuclein seed quantification in synucleinopathies. Brain. 2022;145:584–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Shahnawaz M, Mukherjee A, Pritzkow S, et al. Discriminating alpha-synuclein strains in Parkinson’s disease and multiple system atrophy. Nature. 2020;578:273–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. De Luca CMG, Elia AE, Portaleone SM, et al. Efficient RT-QuIC seeding activity for alpha-synuclein in olfactory mucosa samples of patients with Parkinson’s disease and multiple system atrophy. Transl Neurodegener. 2019;8:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Yoo D, Bang JI, Ahn C, et al. Diagnostic value of alpha-synuclein seeding amplification assays in alpha-synucleinopathies: A systematic review and meta-analysis. Parkinsonism Relat Disord. 2022;104:99–109. [DOI] [PubMed] [Google Scholar]
  • 91. Wang YS, Hu JY, Chen XF, et al. Real-time quaking-induced conversion assay is accurate for Lewy body diseases: A meta-analysis. Neurol Sci. 2022;43:4125–4132. [DOI] [PubMed] [Google Scholar]
  • 92. Peng C, Gathagan RJ, Covell DJ, et al. Cellular milieu imparts distinct pathological alpha-synuclein strains in alpha-synucleinopathies. Nature. 2018;557:558–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Wang Z, Becker K, Donadio V, et al. Skin alpha-synuclein aggregation seeding activity as a novel biomarker for Parkinson disease. JAMA Neurol. 2020;78:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Manne S, Kondru N, Jin H, et al. alpha-Synuclein real-time quaking-induced conversion in the submandibular glands of Parkinson’s disease patients. Mov Disord. 2020;35:268–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Chahine LM, Beach TG, Adler CH, et al. Central and peripheral alpha-synuclein in Parkinson disease detected by seed amplification assay. Ann Clin Transl Neurol. 2023;10:696–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Vivacqua G, Mason M, De Bartolo MI, et al. Salivary alpha-synuclein RT-QuIC correlates with disease severity in de novo Parkinson’s disease. Mov Disord. 2023;38:153–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Okuzumi A, Hatano T, Matsumoto G, et al. Propagative alpha-synuclein seeds as serum biomarkers for synucleinopathies. Nat Med. 2023;29:1448–1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Kluge A, Bunk J, Schaeffer E, et al. Detection of neuron-derived pathological alpha-synuclein in blood. Brain. 2022;145:3058–3071. [DOI] [PubMed] [Google Scholar]
  • 99. Grossauer A, Hemicker G, Krismer F, et al. alpha-Synuclein seed amplification assays in the diagnosis of synucleinopathies using cerebrospinal fluid—A systematic review and meta-analysis. Mov Disord Clin Prac. 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Wang H, Atik A, Stewart T, et al. Plasma alpha-synuclein and cognitive impairment in the Parkinson’s associated risk syndrome: A pilot study. Neurobiol Dis. 2018;116:53–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Majbour NK, Vaikath NN, Eusebi P, et al. Longitudinal changes in CSF alpha-synuclein species reflect Parkinson’s disease progression. Mov Disord. 2016;31:1535–1542. [DOI] [PubMed] [Google Scholar]
  • 102. Lin CH, Liu HC, Yang SY, Yang KC, Wu CC, Chiu MJ. Plasma pS129-alpha-synuclein is a surrogate biofluid marker of motor severity and progression in Parkinson’s disease. J Clin Med. 2019;8(10):1601.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Majbour NK, Abdi IY, Dakna M, et al. Cerebrospinal alpha-synuclein oligomers reflect disease motor severity in DeNoPa longitudinal cohort. Mov Disord. 2021;36:2048–2056. [DOI] [PubMed] [Google Scholar]
  • 104. Imam SZ, Zhou Q, Yamamoto A, et al. Novel regulation of parkin function through c-Abl-mediated tyrosine phosphorylation: Implications for Parkinson’s disease. J Neurosci. 2011;31:157–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Na CH, Sathe G, Rosenthal LS, et al. Development of a novel method for the quantification of tyrosine 39 phosphorylated alpha- and beta-synuclein in human cerebrospinal fluid. Clin Proteomics. 2020;17:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Fernandez E, Garcia-Moreno JM, de Pablos AM, Chacon J. May the evaluation of nitrosative stress through selective increase of 3-nitrotyrosine proteins other than nitroalbumin and dominant tyrosine-125/136 nitrosylation of serum -synuclein serve for diagnosis of sporadic Parkinson’s disease? Antioxid Redox Sign. 2013;19:912–918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Beach TG, Adler CH, Sue LI, et al. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol. 2010;119:689–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Harapan BN, Frydrychowicz C, Classen J, et al. No enhanced (p-) alpha-synuclein deposition in gastrointestinal tissue of Parkinson’s disease patients. Parkinsonism Relat Disord. 2020;80:82–88. [DOI] [PubMed] [Google Scholar]
  • 109. Bu LL, Huang KX, Zheng DZ, et al. Alpha-synuclein accumulation and its phosphorylation in the enteric nervous system of patients without neurodegeneration: An explorative study. Front Aging Neurosci. 2020;12:575481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Murakami H, Tokuda T, El-Agnaf OMA, et al. Correlated levels of cerebrospinal fluid pathogenic proteins in drug-naive Parkinson’s disease. BMC Neurol. 2019;19:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Agliardi C, Meloni M, Guerini FR, et al. Oligomeric alpha-Syn and SNARE complex proteins in peripheral extracellular vesicles of neural origin are biomarkers for Parkinson’s disease. Neurobiol Dis. 2021;148:105185. [DOI] [PubMed] [Google Scholar]
  • 112. Rani K, Mukherjee R, Singh E, et al. Neuronal exosomes in saliva of Parkinson’s disease patients: A pilot study. Parkinsonism Relat Disord. 2019;67:21–23. [DOI] [PubMed] [Google Scholar]
  • 113. Iranzo A, Fairfoul G, Ayudhaya ACN, et al. Detection of alpha-synuclein in CSF by RT-QuIC in patients with isolated rapid-eye-movement sleep behaviour disorder: A longitudinal observational study. Lancet Neurol. 2021;20:203–212. [DOI] [PubMed] [Google Scholar]
  • 114. Chen GF, Xu TH, Yan Y, et al. Amyloid beta: Structure, biology and structure-based therapeutic development. Acta Pharmacol Sin. 2017;38:1205–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Bentahir M, Nyabi O, Verhamme J, et al. Presenilin clinical mutations can affect gamma-secretase activity by different mechanisms. J Neurochem. 2006;96:732–742. [DOI] [PubMed] [Google Scholar]
  • 116. Mietelska-Porowska A, Wasik U, Goras M, Filipek A, Niewiadomska G. Tau protein modifications and interactions: Their role in function and dysfunction. Int J Mol Sci. 2014;15:4671–4713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Irwin DJ, Cohen TJ, Grossman M, et al. Acetylated tau, a novel pathological signature in Alzheimer’s disease and other tauopathies. Brain. 2012;135(Pt 3):807–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Yoshida M. Astrocytic inclusions in progressive supranuclear palsy and corticobasal degeneration. Neuropathology. 2014;34:555–570. [DOI] [PubMed] [Google Scholar]
  • 119. Aerts MB, Esselink RA, Bloem BR, Verbeek MM. Cerebrospinal fluid tau and phosphorylated tau protein are elevated in corticobasal syndrome. Mov Disord. 2011;26:169–173. [DOI] [PubMed] [Google Scholar]
  • 120. Constantinides VC, Paraskevas GP, Emmanouilidou E, et al. CSF Biomarkers beta-amyloid, tau proteins and a-synuclein in the differential diagnosis of Parkinson-plus syndromes. J Neurol Sci. 2017;382:91–95. [DOI] [PubMed] [Google Scholar]
  • 121. Saijo E, Metrick MA, Koga S, et al. 4–Repeat Tau seeds and templating subtypes as brain and CSF biomarkers of frontotemporal lobar degeneration. Acta Neuropathol. 2020;139:79–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Dubois B, Feldman HH, Jacova C, et al. Advancing research diagnostic criteria for Alzheimer’s disease: The IWG-2 criteria. Lancet Neurol. 2014;13:614–629. [DOI] [PubMed] [Google Scholar]
  • 123. Kurata T, Kawarabayashi T, Murakami T, et al. Enhanced accumulation of phosphorylated alpha-synuclein in double transgenic mice expressing mutant beta-amyloid precursor protein and presenilin-1. J Neurosci Res. 2007;85:2246–2252. [DOI] [PubMed] [Google Scholar]
  • 124. Parnetti L, Tiraboschi P, Lanari A, et al. Cerebrospinal fluid biomarkers in Parkinson’s disease with dementia and dementia with Lewy bodies. Biol Psychiatry. 2008;64:850–855. [DOI] [PubMed] [Google Scholar]
  • 125. Clinton LK, Blurton-Jones M, Myczek K, Trojanowski JQ, LaFerla FM. Synergistic interactions between abeta, tau, and alpha-synuclein: Acceleration of neuropathology and cognitive decline. J Neurosci. 2010;30:7281–7289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Lin CH, Yang SY, Horng HE, et al. Plasma biomarkers differentiate Parkinson’s disease from atypical parkinsonism syndromes. Front Aging Neurosci. 2018;10:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Guo JL, Covell DJ, Daniels JP, et al. Distinct alpha-synuclein strains differentially promote tau inclusions in neurons. Cell. 2013;154:103–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Jellinger KA, Seppi K, Wenning GK, Poewe W. Impact of coexistent Alzheimer pathology on the natural history of Parkinson’s disease. J Neural Transm (Vienna). 2002;109:329–339. [DOI] [PubMed] [Google Scholar]
  • 129. Irwin DJ, Lee VM, Trojanowski JQ. Parkinson’s disease dementia: Convergence of alpha-synuclein, tau and amyloid-beta pathologies. Nat Rev Neurosci. 2013;14:626–636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Siderowf A, Xie SX, Hurtig H, et al. CSF Amyloid {beta} 1–42 predicts cognitive decline in Parkinson disease. Neurology. 2010;75:1055–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Alves G, Lange J, Blennow K, et al. CSF Abeta42 predicts early-onset dementia in Parkinson disease. Neurology. 2014;82:1784–1790. [DOI] [PubMed] [Google Scholar]
  • 132. Blennow K, Biscetti L, Eusebi P, Parnetti L. Cerebrospinal fluid biomarkers in Alzheimer’s and Parkinson’s diseases-from pathophysiology to clinical practice. Mov Disord. 2016;31:836–847. [DOI] [PubMed] [Google Scholar]
  • 133. Zetterberg H. Plasma amyloid beta-quo vadis? Neurobiol Aging. 2015;36:2671–2673. [DOI] [PubMed] [Google Scholar]
  • 134. Teunissen CE, Chiu MJ, Yang CC, et al. Plasma amyloid-beta (Abeta42) correlates with cerebrospinal fluid Abeta42 in Alzheimer’s disease. J Alzheimers Dis. 2018;62:1857–1863. [DOI] [PubMed] [Google Scholar]
  • 135. Chojdak-Lukasiewicz J, Malodobra-Mazur M, Zimny A, Noga L, Paradowski B. Plasma tau protein and Abeta42 level as markers of cognitive impairment in patients with Parkinson’s disease. Adv Clin Exp Med. 2020;29:115–121. [DOI] [PubMed] [Google Scholar]
  • 136. Chen NC, Chen HL, Li SH, et al. Plasma levels of alpha-synuclein, Abeta-40 and T-tau as biomarkers to predict cognitive impairment in Parkinson’s disease. Front Aging Neurosci. 2020;12:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Randall J, Mortberg E, Provuncher GK, et al. Tau proteins in serum predict neurological outcome after hypoxic brain injury from cardiac arrest: Results of a pilot study. Resuscitation. 2013;84:351–356. [DOI] [PubMed] [Google Scholar]
  • 138. Lin WT, Shaw JS, Cheng FY, Chen PH. Plasma total tau predicts executive dysfunction in Parkinson’s disease. Acta Neurol Scand. 2022;145:30–37. [DOI] [PubMed] [Google Scholar]
  • 139. Chung CC, Chan L, Chen JH, Bamodu OA, Chiu HW, Hong CT. Plasma extracellular vesicles tau and beta-amyloid as biomarkers of cognitive dysfunction of Parkinson’s disease. FASEB J. 2021;35:e21895. [DOI] [PubMed] [Google Scholar]
  • 140. Chan L, Chung CC, Hsieh YC, Wu RM, Hong CT. Plasma extracellular vesicle tau, beta-amyloid, and alpha-synuclein and the progression of Parkinson’s disease: A follow-up study. Ther Adv Neurol Disord. 2023;16:17562864221150329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Blommer J, Pitcher T, Mustapic M, et al. Extracellular vesicle biomarkers for cognitive impairment in Parkinson’s disease. Brain. 2022;146:195–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Verde F. Tau proteins in blood as biomarkers of Alzheimer’s disease and other proteinopathies. J Neural Transm. 2022;129:239–259. [DOI] [PubMed] [Google Scholar]
  • 143. Batzu L, Rota S, Hye A, et al. Plasma p-tau181, neurofilament light chain and association with cognition in Parkinson’s disease. NPJ Parkinsons Dis. 2022;8:154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Pagonabarraga J, Perez-Gonzalez R, Bejr-Kasem H, et al. Dissociable contribution of plasma NfL and p-tau181 to cognitive impairment in Parkinson’s disease. Parkinsonism Relat Disord. 2022;105:132–138. [DOI] [PubMed] [Google Scholar]
  • 145. Chiu MJ, Yang SY, Chen TF, et al. Synergistic association between plasma Abeta(1–42) and p-tau in Alzheimer’s disease but not in Parkinson’s disease or frontotemporal dementia. Acs Chem Neurosci. 2021;12:1376–1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Ransohoff RM. How neuroinflammation contributes to neurodegeneration. Science. 2016;353:777–783. [DOI] [PubMed] [Google Scholar]
  • 147. Su W, Chen HB, Li SH, Wu DY. Correlational study of the serum levels of the glial fibrillary acidic protein and neurofilament proteins in Parkinson’s disease patients. Clin Neurol Neurosur. 2012;114:372–375. [DOI] [PubMed] [Google Scholar]
  • 148. Oeckl P, Halbgebauer S, Anderl-Straub S, et al. Glial fibrillary acidic protein in serum is increased in Alzheimer’s disease and correlates with cognitive impairment. J Alzheimers Dis. 2019;67:481–488. [DOI] [PubMed] [Google Scholar]
  • 149. Olsson B, Constantinescu R, Holmberg B, Andreasen N, Blennow K, Zetterberg H. The glial marker YKL-40 is decreased in synucleinopathies. Mov Disord. 2013;28:1882–1885. [DOI] [PubMed] [Google Scholar]
  • 150. Wennstrom M, Surova Y, Hall S, et al. The inflammatory marker YKL-40 is elevated in cerebrospinal fluid from patients with Alzheimer’s but not Parkinson’s disease or dementia with Lewy bodies. PLoS One. 2015;10:e0135458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Magdalinou NK, Paterson RW, Schott JM, et al. A panel of nine cerebrospinal fluid biomarkers may identify patients with atypical parkinsonian syndromes. J Neurol Neurosurg Psychiatry. 2015;86:1240–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Tang Y, Han L, Li S, et al. Plasma GFAP in Parkinson’s disease with cognitive impairment and its potential to predict conversion to dementia. NPJ Parkinsons Dis. 2023;9:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Munoz-Delgado L, Macias-Garcia D, Jesus S, et al. Peripheral immune profile and neutrophil-to-lymphocyte ratio in Parkinson’s disease. Mov Disord. 2021;36:2426–2430. [DOI] [PubMed] [Google Scholar]
  • 154. Alvarez-Luquin DD, Arce-Sillas A, Leyva-Hernandez J, et al. Regulatory impairment in untreated Parkinson’s disease is not restricted to Tregs: Other regulatory populations are also involved. J Neuroinflammation. 2019;16:212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Kustrimovic N, Comi C, Magistrelli L, et al. Parkinson’s disease patients have a complex phenotypic and functional Th1 bias: Cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naive and drug-treated patients. J Neuroinflammation. 2018;15:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Rocha NP, Assis F, Scalzo PL, et al. Reduced activated T lymphocytes (CD4+CD25+) and plasma levels of cytokines in Parkinson’s disease. Mol Neurobiol. 2018;55:1488–1497. [DOI] [PubMed] [Google Scholar]
  • 157. Sun C, Zhao Z, Yu W, et al. Abnormal subpopulations of peripheral blood lymphocytes are involved in Parkinson’s disease. Ann Transl Med. 2019;7:637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Sanjari Moghaddam H, Ghazi Sherbaf F, Mojtahed Zadeh M, Ashraf-Ganjouei A, Aarabi MH. Association between peripheral inflammation and DATSCAN data of the striatal nuclei in different motor subtypes of Parkinson disease. Front Neurol. 2018;9:234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Munoz-Delgado L, Labrador-Espinosa MA, Macias-Garcia D, et al. Peripheral inflammation is associated with dopaminergic degeneration in Parkinson’s disease. Mov Disord. 2023;38:755–763. [DOI] [PubMed] [Google Scholar]
  • 160. Magistrelli L, Storelli E, Rasini E, et al. Relationship between circulating CD4+ T lymphocytes and cognitive impairment in patients with Parkinson’s disease. Brain Behav Immun. 2020;89:668–674. [DOI] [PubMed] [Google Scholar]
  • 161. Saunders JA, Estes KA, Kosloski LM, et al. CD4+ regulatory And effector/memory T cell subsets profile motor dysfunction in Parkinson’s disease. J Neuroimmune Pharmacol. 2012;7:927–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Karaaslan Z, Kahraman OT, Sanli E, et al. Inflammation and regulatory T cell genes are differentially expressed in peripheral blood mononuclear cells of Parkinson’s disease patients. Sci Rep. 2021;11:2316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Akil E, Bulut A, Kaplan I, Ozdemir HH, Arslan D, Aluclu MU. The increase of carcinoembryonic antigen (CEA), high-sensitivity C-reactive protein, and neutrophil/lymphocyte ratio in Parkinson’s disease. Neurol Sci. 2015;36:423–428. [DOI] [PubMed] [Google Scholar]
  • 164. Jin H, Gu HY, Mao CJ, Chen J, Liu CF. Association of inflammatory factors and aging in Parkinson’s disease. Neurosci Lett. 2020;736:135259. [DOI] [PubMed] [Google Scholar]
  • 165. Qiu X, Xiao Y, Wu J, Gan L, Huang Y, Wang J. C-reactive protein and risk of Parkinson’s disease: A systematic review and meta-analysis. Front Neurol. 2019;10:384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Vesely B, Dufek M, Thon V, et al. Interleukin 6 and complement serum level study in Parkinson’s disease. J Neural Transm (Vienna). 2018;125:875–881. [DOI] [PubMed] [Google Scholar]
  • 167. Karpenko MN, Vasilishina AA, Gromova EA, Muruzheva ZM, Miliukhina IV, Bernadotte A. Interleukin-1beta, interleukin-1 receptor antagonist, interleukin-6, interleukin-10, and tumor necrosis factor-alpha levels in CSF and serum in relation to the clinical diversity of Parkinson’s disease. Cell Immunol. 2018;327:77–82. [DOI] [PubMed] [Google Scholar]
  • 168. Kim R, Kim HJ, Kim A, et al. Peripheral blood inflammatory markers in early Parkinson’s disease. J Clin Neurosci. 2018;58:30–33. [DOI] [PubMed] [Google Scholar]
  • 169. Dufek M, Hamanova M, Lokaj J, et al. Serum inflammatory biomarkers in Parkinson’s disease. Parkinsonism Relat Disord. 2009;15:318–320. [DOI] [PubMed] [Google Scholar]
  • 170. Kouchaki E, Kakhaki RD, Tamtaji OR, et al. Increased serum levels of TNF-alpha and decreased serum levels of IL-27 in patients with Parkinson disease and their correlation with disease severity. Clin Neurol Neurosurg. 2018;166:76–79. [DOI] [PubMed] [Google Scholar]
  • 171. Wang XM, Zhang YG, Li AL, et al. Relationship between levels of inflammatory cytokines in the peripheral blood and the severity of depression and anxiety in patients with Parkinson’s disease. Eur Rev Med Pharmaco. 2016;20:3853–3856. [PubMed] [Google Scholar]
  • 172. Gupta V, Garg RK, Khattri S. Levels of IL-8 and TNF-alpha decrease in Parkinson’s disease. Neurol Res. 2016;38:98–102. [DOI] [PubMed] [Google Scholar]
  • 173. Lindqvist D, Kaufman E, Brundin L, Hall S, Surova Y, Hansson O. Non-motor symptoms in patients with Parkinson’s disease—Correlations with inflammatory cytokines in serum. PLoS One. 2012;7:e47387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Schroder JB, Pawlowski M, Meyer Zu Horste G, et al. Immune cell activation in the cerebrospinal fluid of patients with Parkinson’s disease. Front Neurol. 2018;9:1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Qin XY, Zhang SP, Cao C, Loh YP, Cheng Y. Aberrations in peripheral inflammatory cytokine levels in Parkinson disease: A systematic review and meta-analysis. JAMA Neurol. 2016;73:1316–1324. [DOI] [PubMed] [Google Scholar]
  • 176. Menza M, Dobkin RD, Marin H, et al. The role of inflammatory cytokines in cognition and other non-motor symptoms of Parkinson’s disease. Psychosomatics. 2010;51:474–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Sawada H, Oeda T, Umemura A, et al. Baseline C-reactive protein levels and life prognosis in Parkinson disease. PLoS One. 2015;10:e0134118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Rentzos M, Nikolaou C, Andreadou E, et al. Circulating interleukin-15 and RANTES chemokine in Parkinson’s disease. Acta Neurol Scand. 2007;116:374–379. [DOI] [PubMed] [Google Scholar]
  • 179. Tang P, Chong L, Li X, et al. Correlation between serum RANTES levels and the severity of Parkinson’s disease. Oxid Med Cell Longev. 2014;2014:208408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Williams-Gray CH, Wijeyekoon R, Yarnall AJ, et al. Serum immune markers and disease progression in an incident Parkinson’s disease cohort (ICICLE-PD). Mov Disord. 2016;31:995–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Rathnayake D, Chang T, Udagama P. Selected serum cytokines and nitric oxide as potential multi-marker biosignature panels for Parkinson disease of varying durations: A case-control study. BMC Neurol. 2019;19:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Kalia LV, Lang AE, Hazrati LN, et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol. 2015;72:100–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Sosero YL, Gan-Or Z. LRRK2 And Parkinson’s disease: From genetics to targeted therapy. Ann Clin Transl Neurol. 2023;10:850–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Brockmann K, Quadalti C, Lerche S, et al. Association between CSF alpha-synuclein seeding activity and genetic status in Parkinson’s disease and dementia with Lewy bodies. Acta Neuropathol Commun. 2021;9:175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Fraser KB, Rawlins AB, Clark RG, et al. Ser(P)-1292 LRRK2 in urinary exosomes is elevated in idiopathic Parkinson’s disease. Mov Disord. 2016;31:1543–1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Wang SJ, Liu ZY, Ye T, et al. Elevated LRRK2 autophosphorylation in brain-derived and peripheral exosomes in LRRK2 mutation carriers. Acta Neuropathol Com. 2017;5:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Jennings D, Huntwork-Rodriguez S, Vissers M, et al. LRRK2 Inhibition by BIIB122 in healthy participants and patients with Parkinson’s disease. Mov Disord. 2023;38:386–398. [DOI] [PubMed] [Google Scholar]
  • 188. Papadimitriou D, Antonelou R, Miligkos M, et al. Motor and nonmotor features of carriers of the p.A53T alpha-synuclein mutation: A longitudinal study. Mov Disord. 2016;31:1226–1230. [DOI] [PubMed] [Google Scholar]
  • 189. Brockmann K, Srulijes K, Pflederer S, et al. GBA-associated Parkinson’s disease: Reduced survival and more rapid progression in a prospective longitudinal study. Mov Disord. 2015;30:407–411. [DOI] [PubMed] [Google Scholar]
  • 190. Brockmann K, Schulte C, Deuschle C, et al. Neurodegenerative CSF markers in genetic and sporadic PD: Classification and prediction in a longitudinal study. Parkinsonism Relat Disord. 2015;21:1427–1434. [DOI] [PubMed] [Google Scholar]
  • 191. Pankratz N, Byder L, Halter C, et al. Presence of an APOE4 allele results in significantly earlier onset of Parkinson’s disease and a higher risk with dementia. Mov Disord. 2006;21:45–49. [DOI] [PubMed] [Google Scholar]
  • 192. Liu JY, Ma LZ, Wang J, et al. Age-related association between APOE varepsilon4 and cognitive progression in de novo Parkinson’s disease. J Alzheimers Dis. 2023;91:1121–1132. [DOI] [PubMed] [Google Scholar]
  • 193. Tan MMX, Lawton MA, Jabbari E, et al. Genome-wide association studies of cognitive and motor progression in Parkinson’s disease. Mov Disord. 2021;36:424–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Winder-Rhodes SE, Evans JR, Ban M, et al. Glucocerebrosidase mutations influence the natural history of Parkinson’s disease in a community-based incident cohort. Brain. 2013;136:392–399. [DOI] [PubMed] [Google Scholar]
  • 195. Davis MY, Johnson CO, Leverenz JB, et al. Association of GBA mutations and the E326K polymorphism with motor and cognitive progression in Parkinson disease. JAMA Neurol. 2016;73:1217–1224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Liu GQ, Boot B, Locascio JJ, et al. Specifically neuropathic Gaucher’s mutations accelerate cognitive decline in Parkinson’s. Ann Neurol. 2016;80:674–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Iwaki H, Blauwendraat C, Leonard HL. Genetic risk of Parkinson disease and progression: An analysis of 13 longitudinal cohorts. Neurol-Genet. 2019;5:e348.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Lawton M, Tan MM, Ben-Shlomo Y, et al. Genetics of validated Parkinson’s disease subtypes in the Oxford discovery and tracking Parkinson’s cohorts. J Neurol Neurosurg Psychiatry. 2022;93:952–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Liu G, Peng J, Liao Z, et al. Genome-wide survival study identifies a novel synaptic locus and polygenic score for cognitive progression in Parkinson’s disease. Nat Genet. 2021;53:787–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Kern F, Krammes L, Danz K, et al. Validation of human microRNA target pathways enables evaluation of target prediction tools. Nucleic Acids Res. 2021;49:127–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Ding H, Huang Z, Chen M, et al. Identification of a panel of five serum miRNAs as a biomarker for Parkinson’s disease. Parkinsonism Relat Disord. 2016;22:68–73. [DOI] [PubMed] [Google Scholar]
  • 202. Caldi Gomes L, Roser AE, Jain G, et al. MicroRNAs from extracellular vesicles as a signature for Parkinson’s disease. Clin Transl Med. 2021;11:e357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Tomlinson PR, Zheng Y, Fischer R, et al. Identification of distinct circulating exosomes in Parkinson’s disease. Ann Clin Transl Neur. 2015;2:353–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Gui YX, Liu H, Zhang LS, Lv W, Hu XY. Altered microRNA profiles in cerebrospinal fluid exosome in Parkinson disease and Alzheimer disease. Oncotarget. 2015;6:37043–37053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. O’Regan G, deSouza RM, Balestrino R, Schapira AH. Glucocerebrosidase mutations in Parkinson disease. J Parkinsons Dis. 2017;7:411–422. [DOI] [PubMed] [Google Scholar]
  • 206. Sevlever D, Jiang PZ, Yen SHC. Cathepsin D is the main lysosomal enzyme involved in the degradation of alpha-synuclein and generation of its carboxy-terminally truncated species. Biochemistry. 2008;47:9678–9687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Lerche S, Schulte C, Wurster I, et al. The mutation matters: CSF profiles of GCase, sphingolipids, alpha-synuclein in PD(GBA). Mov Disord. 2021;36:1216–1228. [DOI] [PubMed] [Google Scholar]
  • 208. Parnetti L, Paciotti S, Eusebi P, et al. Cerebrospinal fluid beta-glucocerebrosidase activity is reduced in Parkinson’s disease patients. Mov Disord. 2017;32:1423–1431. [DOI] [PubMed] [Google Scholar]
  • 209. Omer N, Giladi N, Gurevich T, et al. Glucocerebrosidase activity is not associated with Parkinson’s disease risk or severity. Mov Disord. 2022;37:651–652. [DOI] [PubMed] [Google Scholar]
  • 210. Oftedal L, Maple-Grodem J, Dalen I, et al. Association of CSF glucocerebrosidase activity with the risk of incident dementia in patients with Parkinson disease. Neurology. 2023;100:e388–e395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Atashrazm F, Hammond D, Perera G, et al. Reduced glucocerebrosidase activity in monocytes from patients with Parkinson’s disease. Sci Rep. 2018;8:15446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Alcalay RN, Levy OA, Waters CC, et al. Glucocerebrosidase activity in Parkinson’s disease with and without GBA mutations. Brain. 2015;138:2648–2658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Mielke MM, Maetzler W, Haughey NJ, et al. Plasma ceramide and glucosylceramide metabolism is altered in sporadic Parkinson’s disease and associated with cognitive impairment: A pilot study. PLos One. 2013;8:e73094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. den Heijer JM, Cullen VC, Pereira DR, et al. A biomarker study in patients with GBA1-Parkinson’s disease and healthy controls. Mov Disord. 2023;38;783–795. [DOI] [PubMed] [Google Scholar]
  • 215. Nicklas WJ, Saporito M, Basma A, Geller HM, Heikkila RE. Mitochondrial mechanisms of neurotoxicity. Ann N Y Acad Sci. 1992;648:28–36. [DOI] [PubMed] [Google Scholar]
  • 216. Larsen SB, Hanss Z, Kruger R. The genetic architecture of mitochondrial dysfunction in Parkinson’s disease. Cell Tissue Res. 2018;373:21–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Kahle PJ, Waak J, Gasser T. DJ-1 and prevention of oxidative stress in Parkinson’s disease and other age-related disorders. Free Radic Biol Med. 2009;47:1354–1361. [DOI] [PubMed] [Google Scholar]
  • 218. Waragai M, Wei J, Fujita 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] [PubMed] [Google Scholar]
  • 219. Waragai M, Nakai M, Wei J, 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] [PubMed] [Google Scholar]
  • 220. Salvesen L, Bech S, Lokkegaard A, et al. The DJ-1 concentration in cerebrospinal fluid does not differentiate among Parkinsonian syndromes. Parkinsonism Relat Disord. 2012;18:899–901. [DOI] [PubMed] [Google Scholar]
  • 221. Herbert MK, Eeftens JM, Aerts MB, et al. CSF Levels of DJ-1 and tau distinguish MSA patients from PD patients and controls. Parkinsonism Relat Disord. 2014;20:112–115. [DOI] [PubMed] [Google Scholar]
  • 222. An C, Pu X, Xiao W, Zhang H. Expression of the DJ-1 protein in the serum of Chinese patients with Parkinson’s disease. Neurosci Lett. 2018;665:236–239. [DOI] [PubMed] [Google Scholar]
  • 223. Maita C, Tsuji S, Yabe I, et al. Secretion of DJ-1 into the serum of patients with Parkinson’s disease. Neurosci Lett. 2008;431:86–89. [DOI] [PubMed] [Google Scholar]
  • 224. Shi M, Zabetian CP, Hancock AM, et al. Significance and confounders of peripheral DJ-1 and alpha-synuclein in Parkinson’s disease. Neurosci Lett. 2010;480:78–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Swatek KN, Usher JL, Kueck AF, et al. Insights into ubiquitin chain architecture using Ub-clipping. Nature. 2019;572:533–537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Hou X, Fiesel FC, Truban D, et al. Age- and disease-dependent increase of the mitophagy marker phospho-ubiquitin in normal aging and Lewy body disease. Autophagy. 2018;14:1404–1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Fiesel FC, Ando M, Hudec R, et al. (Patho-)physiological relevance of PINK1-dependent ubiquitin phosphorylation. EMBO Rep. 2015;16:1114–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Piccinin E, Sardanelli AM, Seibel P, Moschetta A, Cocco T, Villani G. PGC-1s in the spotlight with Parkinson’s disease. Int J Mol Sci. 2021;22:3487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Eschbach J, von Einem B, Muller K, et al. Mutual exacerbation of peroxisome proliferator-activated receptor gamma coactivator 1alpha deregulation and alpha-synuclein oligomerization. Ann Neurol. 2015;77:15–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Yang XD, Qian YW, Xu SQ, et al. Expression of the gene coading for PGC-1alpha in peripheral blood leukocytes and related gene variants in patients with Parkinson’s disease. Parkinsonism Relat Disord. 2018;51:30–35. [DOI] [PubMed] [Google Scholar]
  • 231. Yang X, Xu S, Qian Y, He X, Chen S, Xiao Q. Hypermethylation of the gene coding for PGC-1alpha in peripheral blood leukocytes of patients with Parkinson’s disease. Front Neurosci. 2020;14:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Dossi G, Squarcina L, Rango M. In vivo mitochondrial function in idiopathic and genetic Parkinson’s disease. Metabolites. 2019;10:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Davis RL, Wong SL, Carling PJ, Payne T, Sue CM, Bandmann O. Serum FGF-21, GDF-15, and blood mtDNA copy number are not biomarkers of Parkinson disease. Neurol Clin Pract. 2020;10:40–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Chohan H, Senkevich K, Patel RK, et al. Type 2 diabetes as a determinant of Parkinson’s disease risk and progression. Mov Disord. 2021;36:1420–1429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Kotagal V, Albin RL, Muller MLTM, Koeppe RA, Frey KA, Bohnen NI. Diabetes is associated with postural instability and gait difficulty in Parkinson disease. Parkinsonism Relat Disord. 2013;19:522–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Bosco D, Plastino M, Cristiano D, et al. Dementia is associated with insulin resistance in patients with Parkinson’s disease. J Neurol Sci. 2012;315(1–2):39–43. [DOI] [PubMed] [Google Scholar]
  • 237. Athauda D, Evans J, Wernick A, et al. The impact of type 2 diabetes in Parkinson’s disease. Mov Disord. 2022;37:1612–1623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. de Pablo-Fernandez E, Courtney R, Rockliffe A, Gentleman S, Holton JL, Warner TT. Faster disease progression in Parkinson’s disease with type 2 diabetes is not associated with increased alpha-synuclein, tau, amyloid-beta or vascular pathology. Neuropath Appl Neuro. 2021;47:1080–1091. [DOI] [PubMed] [Google Scholar]
  • 239. Gao S, Duan C, Gao G, Wang X, Yang H. Alpha-synuclein overexpression negatively regulates insulin receptor substrate 1 by activating mTORC1/S6K1 signaling. Int J Biochem Cell Biol. 2015;64:25–33. [DOI] [PubMed] [Google Scholar]
  • 240. Athauda D, Foltynie T. Insulin resistance and Parkinson’s disease: A new target for disease modification? Prog Neurobiol. 2016;145–146:98–120. [DOI] [PubMed] [Google Scholar]
  • 241. Tremblay F, Brule S, Hee Um S, et al. Identification of IRS-1 Ser-1101 as a target of S6K1 in nutrient- and obesity-induced insulin resistance. Proc Natl Acad Sci U S A. 2007;104:14056–14061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Bassil F, Delamarre A, Canron MH, et al. Impaired brain insulin signalling in Parkinson’s disease. Neuropathol Appl Neurobiol. 2022;48:e12760. [DOI] [PubMed] [Google Scholar]
  • 243. Bassil F, Canron MH, Vital A, et al. Insulin resistance and exendin-4 treatment for multiple system atrophy. Brain. 2017;140:1420–1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Athauda D, Gulyani S, Karnati HK, et al. Utility of neuronal-derived exosomes to examine molecular mechanisms that affect motor function in patients with Parkinson disease: A secondary analysis of the exenatide-PD trial. JAMA Neurol. 2019;76:420–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Kapogiannis D, Mustapic M, Shardell MD, et al. Association of extracellular vesicle biomarkers with Alzheimer disease in the Baltimore longitudinal study of aging. JAMA Neurol. 2019;76:1340–1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Hogg E, Athreya K, Basile C, Tan EE, Kaminski J, Tagliati M. High prevalence of undiagnosed insulin resistance in non-diabetic subjects with Parkinson’s disease. J Parkinsons Dis. 2018;8:259–265. [DOI] [PubMed] [Google Scholar]
  • 247. Horvath I, Wittung-Stafshede P. Cross-talk between amyloidogenic proteins in type-2 diabetes and Parkinson’s disease. Proc Natl Acad Sci U S A. 2016;113:12473–12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Martinez-Valbuena I, Amat-Villegas I, Valenti-Azcarate R, et al. Interaction of amyloidogenic proteins in pancreatic beta cells from subjects with synucleinopathies. Acta Neuropathol. 2018;135:877–886. [DOI] [PubMed] [Google Scholar]
  • 249. Markaki I, Ntetsika T, Sorjonen K, Svenningsson P; BioPark Study Group . Euglycemia indicates favorable motor outcome in Parkinson’s disease. Mov Disord. 2021;36:1430–1434. [DOI] [PubMed] [Google Scholar]
  • 250. Huxford B, Haque T, Joseph AB, et al. Parkinson’s disease and type 2 diabetes: HbA1c is associated with motor and cognitive severity. Mov Disord. 2022;37:427–428. [DOI] [PubMed] [Google Scholar]
  • 251. Uyar M, Lezius S, Buhmann C, et al. Diabetes, glycated hemoglobin (HbA1c), and neuroaxonal damage in Parkinson’s disease (MARK-PD study). Mov Disord. 2022;37:1299–1304. [DOI] [PubMed] [Google Scholar]
  • 252. Vijiaratnam N, Lawton M, Real R, et al. Diabetes and neuroaxonal damage in Parkinson’s disease. Mov Disord. 2022;37:1568–1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Girges C, Vijiaratnam N, Athauda D, Auld G, Gandhi S, Foltynie T. The future of incretin-based approaches for neurodegenerative diseases in older adults: Which to choose? A review of their potential efficacy and suitability. Drugs Aging. 2021;38:355–373. [DOI] [PubMed] [Google Scholar]
  • 254. Gonzalez AC, Belbin O. Fluid markers of synapse degeneration in synucleinopathies. J Neural Transm. 2022;129:187–206. [DOI] [PubMed] [Google Scholar]
  • 255. Hashimoto M, Rockenstein E, Mante M, Mallory M, Masliah E. beta-Synuclein inhibits alpha-synuclein aggregation: A possible role as an anti-parkinsonian factor. Neuron. 2001;32:213–223. [DOI] [PubMed] [Google Scholar]
  • 256. Oeckl P, Metzger F, Nagl M, et al. Alpha-, beta-, and gamma-synuclein quantification in cerebrospinal fluid by multiple reaction monitoring reveals increased concentrations in Alzheimer’s and Creutzfeldt-Jakob disease but no alteration in synucleinopathies. Mol Cell Proteomics. 2016;15:3126–3138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Halbgebauer S, Oeckl P, Steinacker P, et al. Beta-synuclein in cerebrospinal fluid as an early diagnostic marker of Alzheimer’s disease. J Neurol Neurosur Psychiatry. 2021;92:349–356. [DOI] [PubMed] [Google Scholar]
  • 258. Sjogren M, Minthon L, Davidsson P, et al. CSF Levels of tau, beta-amyloid(1–42) and GAP-43 in frontotemporal dementia, other types of dementia and normal aging. J Neural Transm. 2000;107:563–579. [DOI] [PubMed] [Google Scholar]
  • 259. Remnestal J, Just D, Mitsios N, et al. CSF Profiling of the human brain enriched proteome reveals associations of neuromodulin and neurogranin to Alzheimer’s disease. Proteomics Clin Appl. 2016;10:1242–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Sandelius A, Portelius E, Kallen A, et al. Elevated CSF GAP-43 is Alzheimer’s disease specific and associated with tau and amyloid pathology. Alzheimers Dement. 2019;15:55–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Chatterjee M, van Steenoven I, Huisman E, et al. Contactin-1 is reduced in cerebrospinal fluid of Parkinson’s disease patients and is present within Lewy bodies. Biomolecules. 2020;10(8):1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Enache D, Pereira JB, Jelic V, et al. Increased cerebrospinal fluid concentration of ZnT3 is associated with cognitive impairment in Alzheimer’s disease. J Alzheimers Dis. 2020;77:1143–1155. [DOI] [PubMed] [Google Scholar]
  • 263. Bereczki E, Bogstedt A, Hoglund K, et al. Synaptic proteins in CSF relate to Parkinson’s disease stage markers. NPJ Parkinson Dis. 2017;3:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264. Selnes P, Stav AL, Johansen KK, et al. Impaired synaptic function is linked to cognition in Parkinson’s disease. Ann Clin Transl Neur. 2017;4:700–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Hall S, Janelidze S, Zetterberg H, et al. Cerebrospinal fluid levels of neurogranin in parkinsonian disorders. Mov Disord. 2020;35:513–518. [DOI] [PubMed] [Google Scholar]
  • 266. Portelius E, Olsson B, Hoglund K, et al. Cerebrospinal fluid neurogranin concentration in neurodegeneration: Relation to clinical phenotypes and neuropathology. Acta Neuropathol. 2018;136:363–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Wellington H, Paterson RW, Portelius E, et al. Increased CSF neurogranin concentration is specific to Alzheimer disease. Neurology. 2016;86:829–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Janelidze S, Hertze J, Zetterberg H, et al. Cerebrospinal fluid neurogranin and YKL-40 as biomarkers of Alzheimer’s disease. Ann Clin Transl Neur. 2016;3:12–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. van Steenoven I, Noli B, Cocco C, et al. VGF Peptides in cerebrospinal fluid of patients with dementia with Lewy bodies. Int J Mol Sci. 2019;20:4674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. van Steenoven I, Koel-Simmelink MJA, Vergouw LJM, et al. Identification of novel cerebrospinal fluid biomarker candidates for dementia with Lewy bodies: A proteomic approach. Mol Neurodegener. 2020;15:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Nilsson J, Constantinescu J, Nellgard B, et al. Cerebrospinal fluid biomarkers of synaptic dysfunction are altered in Parkinson’s disease and related disorders. Mov Disord. 2022;38:267–277. [DOI] [PubMed] [Google Scholar]
  • 272. Lerche S, Sjodin S, Brinkmalm A, et al. CSF Protein level of neurotransmitter secretion, synaptic plasticity, and autophagy in PD and DLB. Mov Disord. 2021;36:2595–2604. [DOI] [PubMed] [Google Scholar]
  • 273. Boiten WA, van Steenoven I, Xiao MF, et al. Pathologically decreased CSF levels of synaptic marker NPTX2 in DLB are correlated with levels of alpha-synuclein and VGF. Cells. 2022;11:652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Lewitt PA, Galloway MP, Matson W, et al. Markers of dopamine metabolism in parkinsons-disease. Neurology. 1992;42:2111–2117. [DOI] [PubMed] [Google Scholar]
  • 275. LeWitt P, Schultz L, Auinger P, Lu M; Parkinson Study Group–DATATOP Investigators . CSF Xanthine, homovanillic acid, and their ratio as biomarkers of Parkinson’s disease. Brain Res. 2011;1408:88–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276. Chia LG, Cheng FC, Kuo JS. Monoamines and their metabolites in plasma and lumbar cerebrospinal-fluid of Chinese patients with parkinsons-disease. J Neurol Sci. 1993;116:125–134. [DOI] [PubMed] [Google Scholar]
  • 277. Herbert MK, Kuiperij HB, Bloem BR, Verbeek MM. Levels of HVA, 5-HIAA, and MHPG in the CSF of vascular parkinsonism compared to Parkinson’s disease and controls. J Neurol. 2013;260:3129–3133. [DOI] [PubMed] [Google Scholar]
  • 278. Czech C, Berndt P, Busch K, et al. Metabolite profiling of Alzheimer’s disease cerebrospinal fluid. PLos One. 2012;7:e31501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Eldrup E, Mogensen P, Jacobsen J, Pakkenberg H, Christensen NJ. Csf and plasma-concentrations of free norepinephrine, dopamine, 3,4–dihydroxyphenylacetic acid (Dopac), 3,4-dihydroxyphenylalanine (Dopa), and epinephrine in Parkinsons-disease. Acta Neurol Scand. 1995;92:116–121. [DOI] [PubMed] [Google Scholar]
  • 280. Stefani A, Pierantozzi M, Olivola E, et al. Homovanillic acid in CSF of mild stage Parkinson’s disease patients correlates with motor impairment. Neurochem Int. 2017;105:58–63. [DOI] [PubMed] [Google Scholar]
  • 281. Kremer T, Taylor KI, Siebourg-Polster J, et al. Longitudinal analysis of multiple neurotransmitter metabolites in cerebrospinal fluid in early Parkinson’s disease. Mov Disord. 2021;36:1972–1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Yuan A, Rao MV, Veeranna, Nixon RA. Neurofilaments and neurofilament proteins in health and disease. Cold Spring Harb Perspect Biol. 2017;9:a018309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283. Bridel C, van Wieringen WN, Zetterberg H, et al. Diagnostic value of cerebrospinal fluid neurofilament light protein in neurology: A systematic review and meta-analysis. JAMA Neurol. 2019;76:1035–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Diekamper E, Brix B, Stocker W, et al. Neurofilament levels are reflecting the loss of presynaptic dopamine receptors in movement disorders. Front Neurosci. 2021;15:690013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285. Ye R, Locascio JJ, Goodheart AE, Quan M, Zhang B, Gomperts SN. Serum NFL levels predict progression of motor impairment and reduction in putamen dopamine transporter binding ratios in de novo Parkinson’s disease: An 8-year longitudinal study. Parkinsonism Relat Disord. 2021;85:11–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Gaetani L, Hoglund K, Parnetti L, et al. A new enzyme-linked immunosorbent assay for neurofilament light in cerebrospinal fluid: Analytical validation and clinical evaluation. Alzheimers Res Ther. 2018;10:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Holmberg B, Rosengren L, Karlsson JE, Johnels B. Increased cerebrospinal fluid levels of neurofilament protein in progressive supranuclear palsy and multiple-system atrophy compared with Parkinson’s disease. Mov Disord. 1998;13:70–77. [DOI] [PubMed] [Google Scholar]
  • 288. Hall S, Ohrfelt A, Constantinescu R, et al. Accuracy of a panel of 5 cerebrospinal fluid biomarkers in the differential diagnosis of patients with dementia and/or parkinsonian disorders. Arch Neurol. 2012;69:1445–1452. [DOI] [PubMed] [Google Scholar]
  • 289. Hansson O, Janelidze S, Hall S, et al. Blood-based NfL: A biomarker for differential diagnosis of parkinsonian disorder. Neurology. 2017;88:930–937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290. Wang SY, Chen W, Xu W, et al. Neurofilament light chain in cerebrospinal fluid and blood as a biomarker for neurodegenerative diseases: A systematic review and meta-analysis. J Alzheimers Dis. 2019;72:1353–1361. [DOI] [PubMed] [Google Scholar]
  • 291. Marques TM, van Rumund A, Oeckl P, et al. Serum NFL discriminates Parkinson disease from atypical Parkinsonisms. Neurology. 2019;92:e1479–e1486. [DOI] [PubMed] [Google Scholar]
  • 292. Lin YS, Lee WJ, Wang SJ, Fuh JL. Levels of plasma neurofilament light chain and cognitive function in patients with Alzheimer or Parkinson disease. Sci Rep. 2018;8:17368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293. Lin CH, Li CH, Yang KC, et al. Blood NfL: A biomarker for disease severity and progression in Parkinson disease. Neurology. 2019;93:e1104–e1111. [DOI] [PubMed] [Google Scholar]
  • 294. Mollenhauer B, Dakna M, Kruse N, et al. Validation of serum neurofilament light chain as a biomarker of Parkinson’s disease progression. Mov Disord. 2020;35:1999–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Chung CC, Chan L, Chen JH, Bamodu OA, Hong CT. Neurofilament light chain level in plasma extracellular vesicles and Parkinson’s disease. Ther Adv Neurol Disord. 2020;13:1756286420975917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Vijiaratnam N, Lawton M, Heslegrave AJ, et al. Combining biomarkers for prognostic modelling of Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2022;93:707–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297. Potter-Nerger M, Dutke J, Lezius S, et al. Serum neurofilament light chain and postural instability/gait difficulty (PIGD) subtypes of Parkinson’s disease in the MARK-PD study. J Neural Transm (Vienna). 2022;129:295–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298. Ng ASL, Tan YJ, Yong ACW, et al. Utility of plasma neurofilament light as a diagnostic and prognostic biomarker of the postural instability gait disorder motor subtype in early Parkinson’s disease. Mol Neurodegener. 2020;15:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299. Aamodt WW, Waligorska T, Shen J, et al. Neurofilament light chain as a biomarker for cognitive decline in Parkinson disease. Mov Disord. 2021;36:2945–2950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300. Ma LZ, Zhang C, Wang H, et al. Serum neurofilament dynamics predicts cognitive progression in de novo Parkinson’s disease. J Parkinsons Dis. 2021;11:1117–1127. [DOI] [PubMed] [Google Scholar]
  • 301. Choe CU, Niemann L, Englisch C, et al. Subclinical cardiac microdamage, motor severity, and cognition in Parkinson’s disease. Mov Disord. 2020;35:1863–1868. [DOI] [PubMed] [Google Scholar]
  • 302. Niemann L, Lezius S, Maceski A, et al. Serum neurofilament is associated with motor function, cognitive decline and subclinical cardiac damage in advanced Parkinson’s disease (MARK-PD). Parkinsonism Relat Disord. 2021;90:44–48. [DOI] [PubMed] [Google Scholar]
  • 303. Kim R, Jeon B. Serum neurofilament light chain predicts future freezing of gait in Parkinson’s disease. Parkinsonism Relat Disord. 2021;91:102–104. [DOI] [PubMed] [Google Scholar]
  • 304. Ygland Rodstrom E, Mattsson-Carlgren N, Janelidze S, Hansson O, Puschmann A. Serum neurofilament light chain as a marker of progression in Parkinson’s disease: Long-term observation and implications of clinical subtypes. J Parkinsons Dis. 2022;12:571–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305. Backstrom DC, Eriksson Domellof M, Linder J, et al. Cerebrospinal fluid patterns and the risk of future dementia in early, incident Parkinson disease. JAMA Neurol. 2015;72:1175–1182. [DOI] [PubMed] [Google Scholar]
  • 306. Martinez-Valbuena I, Visanji NP, Olszewska DA, et al. Combining skin alpha-synuclein real-time quaking-induced conversion and circulating neurofilament light chain to distinguish multiple system atrophy and Parkinson’s disease. Mov Disord. 2022;37:648–650. [DOI] [PubMed] [Google Scholar]
  • 307. Compta Y, Painous C, Soto M, et al. Combined CSF alpha-SYN RT-QuIC, CSF NFL and midbrain-pons planimetry in degenerative parkinsonisms: From bedside to bench, and back again. Parkinsonism Relat Disord. 2022;99:33–41. [DOI] [PubMed] [Google Scholar]
  • 308. Gaenslen A, Unmuth B, Godau J, 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] [PubMed] [Google Scholar]
  • 309. Li DH, He YC, Liu J, Chen SD. Diagnostic accuracy of transcranial sonography of the substantia Nigra in Parkinson’s disease: A systematic review and meta-analysis. Sci Rep. 2016;6:20863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310. Berg D, Siefker C, Becker G. Echogenicity of the substantia nigra in Parkinson’s disease and its relation to clinical findings. J Neurol. 2001;248:684–689. [DOI] [PubMed] [Google Scholar]
  • 311. Berg D, Becker G, Zeiler B, et al. Vulnerability of the nigrostriatal system as detected by transcranial ultrasound. Neurology. 1999;53:1026–1031. [DOI] [PubMed] [Google Scholar]
  • 312. Berg D, Merz B, Reiners K, Naumann M, Becker G. Five-year follow-up study of hyperechogenicity of the substantia nigra in Parkinson’s disease. Mov Disord. 2005;20:383–385. [DOI] [PubMed] [Google Scholar]
  • 313. Becker G, Seufert J, Bogdahn U, Reichmann H, Reiners K. Degeneration of substantia nigra in chronic Parkinson’s disease visualized by transcranial color-coded real-time sonography. Neurology. 1995;45:182–184. [DOI] [PubMed] [Google Scholar]
  • 314. Spiegel J, Hellwig D, Mollers MO, et al. Transcranial sonography and [123I]FP-CIT SPECT disclose complementary aspects of Parkinson’s disease. Brain. 2006;129(Pt 5):1188–1193. [DOI] [PubMed] [Google Scholar]
  • 315. Mitchell T, Lehericy S, Chiu SY, Strafella AP, Stoessl AJ, Vaillancourt DE. Emerging neuroimaging biomarkers across disease stage in Parkinson disease: A review. JAMA Neurol. 2021;78:1262–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316. Quattrone A, Bianco MG, Antonini A, et al. Development and validation of automated magnetic resonance Parkinsonism index 2.0 to distinguish progressive supranuclear palsy-Parkinsonism from Parkinson’s disease. Mov Disord. 2022;37:1272–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317. Zeighami Y, Fereshtehnejad SM, Dadar M, Collins DL, Postuma RB, Dagher A. Assessment of a prognostic MRI biomarker in early de novo Parkinson’s disease. Neuroimage Clin. 2019;24:101986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318. Tessa C, Lucetti C, Giannelli M, et al. Progression of brain atrophy in the early stages of Parkinson’s disease: A longitudinal tensor-based morphometry study in de novo patients without cognitive impairment. Hum Brain Mapp. 2014;35:3932–3944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319. Mak E, Su L, Williams GB, et al. Baseline and longitudinal grey matter changes in newly diagnosed Parkinson’s disease: ICICLE-PD study. Brain. 2015;138(Pt 10):2974–2986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Mak E, Su L, Williams GB, et al. Longitudinal whole-brain atrophy and ventricular enlargement in nondemented Parkinson’s disease. Neurobiol Aging. 2017;55:78–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321. Agosta F, Canu E, Stojkovic T, et al. The topography of brain damage at different stages of Parkinson’s disease. Hum Brain Mapp. 2013;34:2798–2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322. Burton EJ, McKeith IG, Burn DJ, Williams ED, O’Brien JT. Cerebral atrophy in Parkinson’s disease with and without dementia: A comparison with Alzheimer’s disease, dementia with Lewy bodies and controls. Brain. 2004;127:791–800. [DOI] [PubMed] [Google Scholar]
  • 323. Hanganu A, Bedetti C, Degroot C, et al. Mild cognitive impairment is linked with faster rate of cortical thinning in patients with Parkinson’s disease longitudinally. Brain. 2014;137:1120–1129. [DOI] [PubMed] [Google Scholar]
  • 324. Sterling NW, Wang M, Zhang LJ, et al. Stage-dependent loss of cortical gyrification as Parkinson disease “unfolds”. Neurology. 2016;86:1143–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325. Lehericy S, Vaillancourt DE, Seppi K, et al. The role of high-field magnetic resonance imaging in parkinsonian disorders: Pushing the boundaries forward. Mov Disord. 2017;32:510–525. [DOI] [PubMed] [Google Scholar]
  • 326. Wolters AF, Heijmans M, Michielse S, et al. The TRACK-PD study: Protocol of a longitudinal ultra-high field imaging study in Parkinson’s disease. BMC Neurol. 2020;20:292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327. Sjöström H, Granberg T, Westman E, Svenningsson P. Quantitative susceptibility mapping differentiates between parkinsonian disorders. Parkinsonism Relat Disord. 2017;44:51–57. [DOI] [PubMed] [Google Scholar]
  • 328. Biondetti E, Gaurav R, Yahia-Cherif L, et al. Spatiotemporal changes in substantia nigra neuromelanin content in Parkinson’s disease. Brain. 2020;143:2757–2770. [DOI] [PubMed] [Google Scholar]
  • 329. Matsuura K, Maeda M, Tabei KI, et al. A longitudinal study of neuromelanin-sensitive magnetic resonance imaging in Parkinson’s disease. Neurosci Lett. 2016;633:112–117. [DOI] [PubMed] [Google Scholar]
  • 330. Castellanos G, Fernandez-Seara MA, Lorenzo-Betancor O, et al. Automated neuromelanin imaging as a diagnostic biomarker for Parkinson’s disease. Mov Disord. 2015;30:945–952. [DOI] [PubMed] [Google Scholar]
  • 331. Gaurav R, Yahia-Cherif L, Pyatigorskaya N, et al. Longitudinal changes in neuromelanin MRI signal in Parkinson’s disease: A progression marker. Mov Disord. 2021;36:1592–1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332. Ohtsuka C, Sasaki M, Konno K, et al. Differentiation of early-stage parkinsonisms using neuromelanin-sensitive magnetic resonance imaging. Parkinsonism Relat Disord. 2014;20:755–760. [DOI] [PubMed] [Google Scholar]
  • 333. Nobileau A, Gaurav R, Chougar L, et al. Neuromelanin-sensitive magnetic resonance imaging changes in the locus coeruleus/subcoeruleus complex in patients with typical and atypical Parkinsonism. Mov Disord. 2023;38:479–484. [DOI] [PubMed] [Google Scholar]
  • 334. Meijer FJA, van Rumund A, Fasen BACM, et al. Susceptibility-weighted imaging improves the diagnostic accuracy of 3T brain MRI in the work-up of Parkinsonism. Am J Neuroradiol. 2015;36:454–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335. Rossi ME, Ruottinen H, Saunamaki T, Elovaara I, Dastidar P. Imaging brain iron and diffusion patterns: A follow-up study of Parkinson’s disease in the initial stages. Acad Radiol. 2014;21:64–71. [DOI] [PubMed] [Google Scholar]
  • 336. Wieler M, Gee M, Martin WRW. Longitudinal midbrain changes in early Parkinson’s disease: Iron content estimated from R-2*/MRI. Parkinsonism Relat Disord. 2015;21:179–183. [DOI] [PubMed] [Google Scholar]
  • 337. Cheng Z, He N, Huang P, et al. Imaging the Nigrosome 1 in the substantia nigra using susceptibility weighted imaging and quantitative susceptibility mapping: An application to Parkinson’s disease. Neuroimage Clin. 2020;25:102103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338. Mahlknecht P, Krismer F, Poewe W, Seppi K. Meta-analysis of dorsolateral nigral hyperintensity on magnetic resonance imaging as a marker for Parkinson’s disease. Mov Disord. 2017;32:619–623. [DOI] [PubMed] [Google Scholar]
  • 339. Du G, Lewis MM, Sica C, et al. Distinct progression pattern of susceptibility MRI in the substantia nigra of Parkinson’s patients. Mov Disord. 2018;33:1423–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340. Hopes L, Grolez G, Moreau C, et al. Magnetic resonance imaging features of the nigrostriatal system: Biomarkers of Parkinson’s disease stages? PLos One. 2016;11:e0147947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341. Ulla M, Bonny JM, Ouchchane L, Rieu I, Claise B, Durif F. Is R2* a new MRI biomarker for the progression of Parkinson’s disease? A longitudinal follow-up. PLoS One. 2013;8:e57904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342. Rolheiser TM, Fulton HG, Good KP, et al. Diffusion tensor imaging and olfactory identification testing in early-stage Parkinson’s disease. J Neurol. 2011;258:1254–1260. [DOI] [PubMed] [Google Scholar]
  • 343. Vaillancourt DE, Spraker MB, Prodoehl J, et al. High-resolution diffusion tensor imaging in the substantia nigra of de novo Parkinson disease. Neurology. 2009;72:1378–1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344. Du G, Lewis MM, Sen S, et al. Imaging nigral pathology and clinical progression in Parkinson’s disease. Mov Disord. 2012;27:1636–1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345. Schwarz ST, Abaei M, Gontu V, Morgan PS, Bajaj N, Auer DP. Diffusion tensor imaging of nigral degeneration in Parkinson’s disease: A region-of-interest and voxel-based study at 3T and systematic review with meta-analysis. Neuroimage Clin. 2013;3:481–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346. Schulz J, Pagano G, Fernandez Bonfante JA, Wilson H, Politis M. Nucleus basalis of meynert degeneration precedes and predicts cognitive impairment in Parkinson’s disease. Brain. 2018;141:1501–1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347. Ofori E, Pasternak O, Planetta PJ, et al. Increased free water in the substantia nigra of Parkinson’s disease: A single-site and multi-site study. Neurobiol Aging. 2015;36:1097–1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348. Burciu RG, Ofori E, Archer DB, et al. Progression marker of Parkinson’s disease: A 4-year multi-site imaging study. Brain. 2017;140:2183–2192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349. Guttuso T Jr, Bergsland N, Hagemeier J, Lichter DG, Pasternak O, Zivadinov R. Substantia Nigra free water increases longitudinally in Parkinson disease. AJNR Am J Neuroradiol. 2018;39:479–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350. Archer DB, Bricker JT, Chu WT, et al. Development and validation of the automated imaging differentiation in Parkinsonism (AID-P): A multi-site machine learning study. Lancet Digit Health. 2019;1:e222–e231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351. Archer DB, Mitchell T, Burciu RG, et al. Magnetic resonance imaging and neurofilament light in the differentiation of Parkinsonism. Mov Disord. 2020;35:1388–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352. Guan J, Rong Y, Wen Y, et al. Detection and application of neurochemical profile by multiple regional (1)H-MRS in Parkinson’s disease. Brain Behav. 2017;7:e00792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353. Cao H, Shi J, Cao B, Kang B, Zhang M, Qu Q. Evaluation of the braak staging of brain pathology with (1)H-MRS in patients with Parkinson’s disease. Neurosci Lett. 2017;660:57–62. [DOI] [PubMed] [Google Scholar]
  • 354. Tsuda M, Asano S, Kato Y, Murai K, Miyazaki M. Differential diagnosis of multiple system atrophy with predominant parkinsonism and Parkinson’s disease using neural networks. J Neurol Sci. 2019;401:19–26. [DOI] [PubMed] [Google Scholar]
  • 355. Taylor-Robinson SD, Turjanski N, Bhattacharya S, et al. A proton magnetic resonance spectroscopy study of the striatum and cerebral cortex in Parkinson’s disease. Metab Brain Dis. 1999;14:45–55. [DOI] [PubMed] [Google Scholar]
  • 356. Camicioli RM, Hanstock CC, Bouchard TP, Gee M, Fisher NJ, Martin WR. Magnetic resonance spectroscopic evidence for presupplementary motor area neuronal dysfunction in Parkinson’s disease. Mov Disord. 2007;22:382–386. [DOI] [PubMed] [Google Scholar]
  • 357. Firbank MJ, Harrison RM, O’Brien JT. A comprehensive review of proton magnetic resonance spectroscopy studies in dementia and Parkinson’s disease. Dement Geriatr Cogn Disord. 2002;14:64–76. [DOI] [PubMed] [Google Scholar]
  • 358. Groger A, Bender B, Wurster I, Chadzynski GL, Klose U, Berg D. Differentiation between idiopathic and atypical parkinsonian syndromes using three-dimensional magnetic resonance spectroscopic imaging. J Neurol Neurosurg Psychiatry. 2013;84:644–649. [DOI] [PubMed] [Google Scholar]
  • 359. Iles RA, Stevens AN, Griffiths JR, Morris PG. Phosphorylation status of liver by 31P-n.m.r. spectroscopy, and its implications for metabolic control. A comparison of 31P–n.m.r. spectroscopy (in vivo and in vitro) with chemical and enzymic determinations of ATP, ADP and Pi. Biochem J. 1985;229:141–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360. Hattingen E, Magerkurth J, Pilatus U, et al. Phosphorus and proton magnetic resonance spectroscopy demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease. Brain. 2009;132(Pt 12):3285–3297. [DOI] [PubMed] [Google Scholar]
  • 361. Hu MT, Taylor-Robinson SD, Chaudhuri KR, et al. Cortical dysfunction in non-demented Parkinson’s disease patients: A combined (31)P-MRS and (18)FDG-PET study. Brain. 2000;123(Pt 2):340–352. [DOI] [PubMed] [Google Scholar]
  • 362. Prasuhn J, Gottlich M, Ebeling B, et al. Assessment of bioenergetic deficits in patients with Parkinson disease and progressive supranuclear palsy using 31P-MRSI. Neurology. 2022;99:e2683–e2692. [DOI] [PubMed] [Google Scholar]
  • 363. Payne T, Appleby M, Buckley E, et al. A double-blind, randomized, placebo-controlled trial of ursodeoxycholic acid (UDCA) in Parkinson’s disease. Mov Disord. 2023;38:1493–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364. Hacker CD, Perlmutter JS, Criswell SR, Ances BM, Snyder AZ. Resting state functional connectivity of the striatum in Parkinson’s disease. Brain. 2012;135(12):3699–3711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365. Szewczyk-Krolikowski K, Menke RA, Rolinski M, et al. Functional connectivity in the basal ganglia network differentiates PD patients from controls. Neurology. 2014;83:208–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366. Baggio HC, Abos A, Segura B, et al. Cerebellar resting-state functional connectivity in Parkinson’s disease and multiple system atrophy: Characterization of abnormalities and potential for differential diagnosis at the single-patient level. Neuroimage Clin. 2019;22:101720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367. Burciu RG, Chung JW, Shukla P, et al. Functional MRI of disease progression in Parkinson disease and atypical parkinsonian syndromes. Neurology. 2016;87:709–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368. Black KJ, Acevedo HK, Koller JM. Dopamine buffering capacity imaging: A pharmacodynamic fMRI method for staging Parkinson disease. Front Neurol. 2020;11:370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369. Eberling JL, Dave KD, Frasier MA. alpha-Synuclein imaging: A critical need for Parkinson’s disease research. J Parkinsons Dis. 2013;3:565–567. [DOI] [PubMed] [Google Scholar]
  • 370. Alzghool OM, van Dongen G, van de Giessen E, Schoonmade L, Beaino W. alpha-Synuclein radiotracer development and in vivo imaging: Recent advancements and new perspectives. Mov Disord. 2022;37:936–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371. Smith R. Initial clinical scans using [18F]ACI-12589, a novel α-synuclein PET-tracer. Alzheimers Dement. 2022;18:e065394. [Google Scholar]
  • 372. Hutchison RM, Evans KC, Fox T, et al. Evaluating dopamine transporter imaging as an enrichment biomarker in a phase 2 Parkinson’s disease trial. BMC Neurol. 2021;21:459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 373. Nicastro N, Garibotto V, Badoud S, Burkhard PR. Scan without evidence of dopaminergic deficit: A 10-year retrospective study. Parkinsonism Relat Disord. 2016;31:53–58. [DOI] [PubMed] [Google Scholar]
  • 374. Benamer TS, Patterson J, Grosset DG, 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]
  • 375. Hong JY, Sunwoo MK, Oh JS, Kim JS, Sohn YH, Lee PH. Persistent drug-induced parkinsonism in patients with normal dopamine transporter imaging. PLoS One. 2016;11:e0157410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376. Palermo G, Giannoni S, Bellini G, Siciliano G, Ceravolo R. Dopamine transporter imaging, current status of a potential biomarker: A comprehensive review. Int J Mol Sci. 2021;22:11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377. Bruck A, Aalto S, Rauhala E, Bergman J, Marttila R, Rinne JO. A follow-up study on 6-[18F]fluoro-L-dopa uptake in early Parkinson’s disease shows nonlinear progression in the putamen. Mov Disord. 2009;24:1009–1015. [DOI] [PubMed] [Google Scholar]
  • 378. Simuni T, Siderowf A, Lasch S, et al. Longitudinal change of clinical and biological measures in early Parkinson’s disease: Parkinson’s progression markers initiative cohort. Mov Disord. 2018;33:771–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379. Nandhagopal R, Kuramoto L, Schulzer M, et al. Longitudinal evolution of compensatory changes in striatal dopamine processing in Parkinson’s disease. Brain. 2011;134:3290–3298. [DOI] [PubMed] [Google Scholar]
  • 380. Lee CS, Samii A, Sossi V, 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]
  • 381. Perlmutter JS, Norris SA. Neuroimaging biomarkers for Parkinson disease: Facts and fantasy. Ann Neurol. 2014;76:769–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382. Vander Borght T, Kilbourn M, Desmond T, Kuhl D, Frey K. The vesicular monoamine transporter is not regulated by dopaminergic drug treatments. Eur J Pharmacol. 1995;294:577–583. [DOI] [PubMed] [Google Scholar]
  • 383. Stoessl AJ, Lehericy S, Strafella AP. Imaging insights into basal ganglia function, Parkinson’s disease, and dystonia. Lancet. 2014;384:532–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384. Palfi S, Gurruchaga JM, Ralph GS, et al. Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson’s disease: A dose escalation, open-label, phase 1/2 trial. Lancet. 2014;383:1138–1146. [DOI] [PubMed] [Google Scholar]
  • 385. Politis M, Wu K, Loane C, et al. Staging of serotonergic dysfunction in Parkinson’s disease: An in vivo C-11-DASB PET study. Neurobiol Dis. 2010;40:216–221. [DOI] [PubMed] [Google Scholar]
  • 386. Fu JF, Matarazzo M, McKenzie J, et al. Serotonergic system impacts levodopa response in early Parkinson’s and future risk of dyskinesia. Mov Disord. 2021;36:389–397. [DOI] [PubMed] [Google Scholar]
  • 387. Kotagal V, Spino C, Bohnen NI, Koeppe R, Albin RL. Serotonin, beta-amyloid, and cognition in Parkinson disease. Ann Neurol. 2018;83:994–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388. Shimada H, Hirano S, Shinotoh H, et al. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET. Neurology. 2009;73:273–278. [DOI] [PubMed] [Google Scholar]
  • 389. Sommerauer M, Fedorova TD, Hansen AK, et al. Evaluation of the noradrenergic system in Parkinson’s disease: An 11C-MeNER PET and neuromelanin MRI study. Brain. 2018;141:496–504. [DOI] [PubMed] [Google Scholar]
  • 390. Matuskey D, Tinaz S, Wilcox KC, et al. Synaptic changes in Parkinson disease assessed with in vivo imaging. Ann Neurol. 2020;87:329–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391. Wilson H, Pagano G, de Natale ER, et al. Mitochondrial complex 1, sigma 1, and synaptic vesicle 2A in early drug-naive Parkinson’s disease. Mov Disord. 2020;35:1416–1427. [DOI] [PubMed] [Google Scholar]
  • 392. Delva A, Van Weehaeghe D, Koole M, Van Laere K, Vandenberghe W. Loss of presynaptic terminal integrity in the substantia nigra in early Parkinson’s disease. Mov Disord. 2020;35:1977–1986. [DOI] [PubMed] [Google Scholar]
  • 393. Andersen KB, Hansen AK, Damholdt MF, et al. Reduced synaptic density in patients with Lewy body dementia: An [(11) C]UCB-J PET imaging study. Mov Disord. 2021;36:2057–2065. [DOI] [PubMed] [Google Scholar]
  • 394. Delva A, Van Laere K, Vandenberghe W. Longitudinal positron emission tomography imaging of presynaptic terminals in early Parkinson’s disease. Mov Disord. 2022;37:1883–1892. [DOI] [PubMed] [Google Scholar]
  • 395. Eckert T, Barnes A, Dhawan V, et al. FDG PET in the differential diagnosis of parkinsonian disorders. Neuroimage. 2005;26:912–921. [DOI] [PubMed] [Google Scholar]
  • 396. Tripathi M, Dhawan V, Peng S, et al. Differential diagnosis of parkinsonian syndromes using F-18 fluorodeoxyglucose positron emission tomography. Neuroradiology. 2013;55:483–492. [DOI] [PubMed] [Google Scholar]
  • 397. Albrecht F, Ballarini T, Neumann J, Schroeter ML. FDG-PET hypometabolism is more sensitive than MRI atrophy in Parkinson’s disease: A whole-brain multimodal imaging meta-analysis. Neuroimage Clin. 2019;21:101594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398. Schindlbeck KA, Lucas-Jimenez O, Tang CC, et al. Metabolic network abnormalities in drug-naive Parkinson’s disease. Mov Disord. 2020;35:587–594. [DOI] [PubMed] [Google Scholar]
  • 399. Tang CC, Poston KL, Eckert T, et al. Differential diagnosis of parkinsonism: A metabolic imaging study using pattern analysis. Lancet Neurol. 2010;9:149–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400. Huang C, Tang C, Feigin A, et al. Changes in network activity with the progression of Parkinson’s disease. Brain. 2007;130(7):1834–1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401. Asanuma K, Tang C, Ma Y, et al. Network modulation in the treatment of Parkinson’s disease. Brain. 2006;129(10):2667–2678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402. Eidelberg D. Metabolic brain networks in neurodegenerative disorders: A functional imaging approach. Trends Neurosci. 2009;32:548–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403. Mattis PJ, Niethammer M, Sako W, et al. Distinct brain networks underlie cognitive dysfunction in Parkinson and Alzheimer diseases. Neurology. 2016;87:1925–1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404. Niethammer M, Eidelberg D. Metabolic brain networks in translational neurology: Concepts and applications. Ann Neurol. 2012;72:635–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 405. Huang C, Mattis P, Tang C, Perrine K, Carbon M, Eidelberg D. Metabolic brain networks associated with cognitive function in Parkinson’s disease. Neuroimage. 2007;34:714–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406. Mizrahi R, Rusjan PM, Kennedy J, et al. Translocator protein (18kDa) polymorphism (rs6971) explains in-vivo brain binding affinity of the PET radioligand [(18)F]-FEPPA. J Cereb Blood Flow Metab. 2012;32:968–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407. Ouchi Y, Yoshikawa E, Sekine Y, et al. Microglial activation and dopamine terminal loss in early Parkinson’s disease. Ann Neurol. 2005;57:168–175. [DOI] [PubMed] [Google Scholar]
  • 408. Ghadery C, Koshimori Y, Coakeley S, et al. Microglial activation in Parkinson’s disease using [(18)F]-FEPPA. J Neuroinflammation. 2017;14:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409. Saeed U, Lang AE, Masellis M. Neuroimaging advances in Parkinson’s disease and atypical Parkinsonian syndromes. Front Neurol. 2020;11:572976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410. Marek K, Siderowf A, Coffey C, et al. Path to prevention (P2P)—Developing a prodromal PD progression biomarker program. Mov Disord. 2019;34:S64–S64. [Google Scholar]
  • 411. Cooper O, Seo H, Andrabi S, et al. Pharmacological rescue of mitochondrial deficits in iPSC-derived neural cells from patients with familial Parkinson’s disease. Sci Transl Med. 2012;4:141ra90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 412. Mullin S, Smith L, Lee K, et al. Ambroxol for the treatment of patients with Parkinson disease with and without glucocerebrosidase gene mutations: A nonrandomized, noncontrolled trial. JAMA Neurol. 2020;77:427–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413. Muller M, Cosman J, Adams J, et al. A progress update on the critical path for Parkinson’s consortium’s pre-competitive 3DT initiative. Mov Disord. 2022;37:S196–S196. [Google Scholar]
  • 414. Tagliapietra M. Aducanumab for the treatment of Alzheimer’s disease. Drugs Today (Barc). 2022;58:465–477. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

awad265_Supplementary_Data

Articles from Brain are provided here courtesy of Oxford University Press

RESOURCES