Abstract
Accurate, reliable, and objective biomarkers for Alzheimer’s disease (AD), Parkinson’s disease (PD), and related age-associated neurodegenerative disorders are urgently needed to assist in both diagnosis, particularly at early stages, and monitoring of disease progression. Technological advancements in protein detection platforms over the last few decades have resulted in a plethora of reported molecular biomarker candidates for both AD and PD; however, very few of these candidates are developed beyond the discovery phase of the biomarker discovery pipeline, a reflection of the current bottleneck within the field. In this review, the expanded use of selected reaction monitoring (SRM) targeted mass spectrometry will be discussed in detail as a platform for systematic verification of large panels of protein biomarker candidates prior to costly validation testing. We also advocate for the coupling of discovery-based proteomics with modern targeted-MS based approaches (e.g. SRM) within a single study in future workflows to expedite biomarker development and validation for AD and PD. It is our hope that improving the efficiency within the biomarker development process by use of an SRM pipeline may ultimately hasten the development of biomarkers that both decrease misdiagnosis of AD and PD and ultimately lead to detection at early stages of disease and objective assessment of disease progression.
Graphical Abstract

Accurate, reliable, and objective biomarkers are urgently needed to assist in both early disease diagnosis and monitoring of disease progression for Alzheimer’s disease (AD) and Parkinson’s disease (PD). Technological developments have resulted in a plethora of biomarker candidates, yet very few become clinically useful. In this review, we highlight the application of selected reaction monitoring (SRM) targeted mass spectrometry as an effective platform for decreasing the bottleneck between biomarker discovery and clinical validation.
Introduction
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are progressive, age-related neurodegenerative disorders with substantial impacts on society (Chan et al. 2016, Coon & Edgerly 1999, Findley 2007). The two hallmark pathologies of AD are β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) (reviewed in (Masters et al. 2015)). Aβ peptides, derived from the amyloid precursor protein (APP), are deposited in neuritic plaques (Cummings & Cole 2002) found in the brains of AD patients and appear to be central to AD pathogenesis. Tau protein is also crucially involved in AD pathogenesis: hyper phosphorylation, truncation, and oligomerization of tau are critical in the formation of NFTs (Cummings & Cole 2002, Semla 2007, Alonso et al. 2001, Alonso et al. 1996, Basurto-Islas et al. 2008, de Calignon et al. 2010). Lewy bodies, the defining pathological characteristic of Lewy body diseases including PD, contain α-synuclein (α-syn) (Kalia & Kalia 2015), particularly modified (e.g., phosphorylated) forms, the role of which in PD pathogenesis is also highlighted by the genetic link between α-syn and PD (Klein & Westenberger 2012). Additionally, they are associated with damage to multiple types of neurons, including dopaminergic neurons (DA) in the substantia nigra (SN) (Poewe et al. 2017, Braak et al. 2003, Halliday et al. 2011). Currently, there is no cure for either disease, and their diagnosis, which requires a combination of clinical assessments, neuropsychological testing, imaging, and exclusion of other neurological disorders (Dubois et al. 2007, Tolosa et al. 2006), most commonly occurs when symptoms develop during later stages of disease progression. Unfortunately, symptoms often do not present before significant, irreversible damage has already occurred in the brain (more than 50% of hippocampal neurons (West et al. 2004, Price et al. 2001) for AD and 60–80% of DA neurons within the SN for PD (Miller & O’Callaghan 2015, Cheng et al. 2010) have already been damaged or lost), resulting in poor prognosis. Additionally, current therapies are primarily focused on alleviating symptoms (cognitive for AD, motor-based for early PD, though cognitive decline is common with progression of PD), but no therapies capable of altering the course of disease exist (Olanow & Stocchi 2017) (Cummings & Cole 2002, Semla 2007). Moreover, objective analysis and monitoring of disease progression or treatment effects is complicated by the lack of suitable markers. Therefore, improvements are urgently needed for diagnosis and progression of AD and PD, particularly at the earliest stages where disease modifying drugs are likely to be more effective, such as through the development of more accurate and accessible biomarkers.
Biomarkers for AD and PD
Advantages of protein biomarkers
Biomarkers are biological characteristics that can be objectively measured to evaluate normal or pathological status of biological systems (Biomarkers Definitions Working 2001, Strimbu & Tavel 2010). Ideally, efficient biomarkers can 1) reveal characteristics of risk, presence, and state of a particular disease (Poste 2011, Puntmann 2009, Zhang et al. 2007, Rifai et al. 2006, Mayeux 2004), 2) be used clinically to predict, diagnose, and monitor disease progression, and 3) assess stage, response to treatment, or prognosis (McDermott et al. 2013). Among the best biomarkers available for neurodegenerative diseases are alterations detected by neuroimaging techniques such as magnetic resonance imaging (MRI) and positron emission tomography (PET) (Ballard et al. 2011, Ghidoni et al. 2013, Risacher & Saykin 2013). For example, both structural and functional neuroimaging have shown promising results in identifying patients with mild cognitive impairment (MCI) who are at a high risk of developing AD (Jack et al. 1999, Wang et al. 2006, Desikan et al. 2010, Forsberg et al. 2008, Koivunen et al. 2010, Hampel et al. 2012), while PET imaging of amyloid burden has potentially improved accuracy of AD diagnosis (Beach et al. 2014, Boccardi et al. 2016). Similarly, PET imaging is quite effective in detecting nigrostriatal degeneration at pre-motor stages of PD (Tzoulis et al. 2013, Nurmi et al. 2001, Broussolle et al. 1999). Unfortunately for both AD and PD, imaging techniques are associated with relatively high costs to healthcare providers and patients, and access to these tools is generally limited to large medical centers (Blennow et al. 2010, Hampel et al. 2008, Hampel et al. 2012, Piccini & Whone 2004), making early diagnostic screening as well as routine monitoring by these tools difficult (Loane & Politis 2011, Schilling et al. 2016). Instead, biomarkers based on differential expression of DNA (Anderson & Kodukula 2014, Podlesniy et al. 2013, Ziegler et al. 2012), RNA (Jeromin & Bowser 2017, Shah et al. 2017, Sheinerman et al. 2017), or proteins (Weiner et al. 2012, Blennow et al. 2010, Hampel et al. 2010, Trojanowski et al. 2010) can be used to determine risk or diagnosis of the disease, while functionally related differences, such as post-translational modifications (PTMs) (Barrett & Timothy Greenamyre 2015), have potential as prognostic predictors of disease progression (Cooper et al. 2006, Breydo et al. 2012, Ferreon & Deniz 2007, Hartl 2017, Sweeney et al. 2017, Robinson 2008). Proteins, which are the major functional units in biological and physiological events, are highly abundant in both tissue and biological fluids (e.g. cerebral spinal fluid [CSF] and blood) and can be detected rather well using protein assays established over the last several decades (Costa et al. 2018). Further, aggregated forms of pathological proteins in both AD and PD are secreted into body fluids and thought to be indicative of pathological development (Musiek & Holtzman 2015, Jouanne et al. 2017, Giraldez-Perez et al. 2014). Thus, protein-based biomarkers have become the most practical and well-studied candidates for molecular biomarkers of AD and PD.
Current protein biomarkers for AD
Currently, amyloid beta (Aβ) and tau proteins/peptides within the CSF comprise the only clinically validated biomarkers for AD (see (Olsson et al. 2016) for a systemic review of these markers). In the CSF, increased total tau or phosphorylated tau protein along with decreased Aβ1–42 (Aβ42) has been demonstrated to provide both high sensitivity and specificity in differentiating AD from controls (Hulstaert et al. 1999, Blennow et al. 2010, Tang & Kumar 2008). Additionally, it has been shown that the combination of total tau and Aβ42 perform well in predicting the conversion of patients with prodromal AD (MCI) to full AD dementia (Hansson et al. 2006, Mattsson et al. 2009, Hertze et al. 2010, Shaw et al. 2009) and also future cognitive decline in asymptomatic older adults (Soldan et al. 2016, Fagan et al. 2007). Additionally, the Aβ42/ Aβ40 ratio seems to show a higher concordance with amyloid load in the brain as assessed by PET than Aβ42 alone (Niemantsverdriet et al. 2017) (Janelidze et al. 2016b) (Lewczuk et al. 2017) and may be of particular use when CSF results are ambiguous (Sauvee et al. 2014). Still, the efficacy of these markers in the CSF during these stages remains to be validated, and, while Aβ42 and tau have demonstrated clinical utility for AD diagnosis, limitations remain for both early detection and differential diagnosis of various dementia and monitoring AD progression (Ritchie et al. 2017).
Current biomarkers for PD
Like Aβ and tau for AD, α-syn, a critical protein involved in PD pathogenesis (Kalia & Kalia 2015), has been extensively evaluated as a likely candidate biomarker for PD given its known involvement within PD pathology, and has shown some promise (Tokuda et al. 2006, Mollenhauer et al. 2011, Parnetti et al. 2011, Wang et al. 2012, Hong et al. 2010, Parnetti et al. 2014, Hall et al. 2015, Korff et al. 2013, Stewart et al. 2015, Fairfoul et al. 2016, Shahnawaz et al. 2017, Groveman et al. 2018). Unfortunately, unlike AD pathological proteins, CSF α-syn has not yet been clinically validated as a biomarker for PD. Various studies have demonstrated that total α-syn protein appears generally lower in CSF of PD patients than compared to healthy controls (Tokuda et al. 2006, Mollenhauer et al. 2011, Parnetti et al. 2011, Wang et al. 2012, Hong et al. 2010, Parnetti et al. 2014, Hall et al. 2015). More recently, α-syn also appears clearly linked to other diseases, including AD (Mollenhauer et al. 2011, Korff et al. 2013). Modified α-syn, including phosphorylated and aggregated forms, have been reported to be more promising candidates (Stewart et al. 2015, Fairfoul et al. 2016, Shahnawaz et al. 2017, Groveman et al. 2018) though further validation is still needed. In addition to α-syn, the AD biomarker tau has become another attractive candidate biomarker for PD, as studies have implicated its association with PD risk and participation in PD pathogenesis based on genome-wide association studies (Zabetian et al. 2007, Simon-Sanchez et al. 2009, Healy et al. 2004, Zhang et al. 2005). Its levels have been reported to be decreased in the CSF of PD patients; however, conflicting studies, showing no change in tau levels, also exist (Kang et al. 2013, Mollenhauer et al. 2011, Hall et al. 2012, Shi et al. 2011, Montine et al. 2010, Abdo et al. 2007). As with tau, the AD marker Aβ42 has also been a useful tool for PD, as studies have reported an association between reduced CSF Aβ42 and cognitive decline in PD (Montine et al. 2010, Skogseth et al. 2015, Terrelonge et al. 2016, Siderowf et al. 2010, Compta et al. 2009). Lastly, DJ-1, another protein that has been associated with an early-onset recessive form of PD (Bonifati et al. 2003), has also generated interest as a candidate marker. However, researchers have struggled to demonstrate consistency between studies (Hong et al. 2010, Waragai et al. 2006).
Additional limitations related to current AD and PD biomarkers
Another limitation related to CSF biomarkers, whether for AD or PD, is the fact that CSF is not suitable for routine clinical practice, as collection requires a lumbar puncture procedure which is often perceived as invasive and painful to patients. As such, patients with minimal or no discernible clinical symptoms are often unwilling to undergo CSF collection, which severely limits access to the prodromal or preclinical population. Thus, both fields would benefit from a collective transition to testing within more clinically-practical biological sources such as blood, which is routinely collected in clinical settings. However, robust diagnostic or predictive blood markers for AD remain to be defined (Hampel et al. 2012, Song et al. 2009, Blennow & Zetterberg 2015, Henriksen et al. 2014), with conflicting findings of minimally decreased or unchanged levels of Aβ42 in AD vs controls (Hampel et al. 2012, Song et al. 2009, Blennow & Zetterberg 2015, Henriksen et al. 2014, Janelidze et al. 2016a) and difficult detection of tau in MCI and AD (Henriksen et al. 2014) without expensive detection platforms (Shi et al. 2016b, Zetterberg et al. 2013). One recent development, reported by two independent studies, is the significantly different Aβ42/ Aβ40 ratio among amyloid positive vs negative patients which may provide improve diagnosis for patients with ambiguous CSF Aβ42 or tau results, which has sparked some hope back into plasma-derived biomarked development for AD (Ovod et al. 2017, Nakamura et al. 2018). Reports on α-syn and other candidates for PD markers in blood are even less consistent and promising (Shi et al. 2016a, Pan et al. 2014, Maita et al. 2008, Waragai et al. 2007, An et al. 2018, Shi et al. 2010, Duran et al. 2010)(El-Agnaf et al. 2006, Lee et al. 2006, Li et al. 2007, Maita et al. 2008, Waragai et al. 2007).
Recent studies by our lab and other groups have suggested blood based but central nervous system (CNS)-specific extracellular vesicles (EVs) might be another alternative biomarker source for neurodegenerative diseases (Howitt & Hill 2016, Malm et al. 2016, Goetzl et al. 2018, Zheng et al. 2017). EVs, which include exosomes and other small membrane-contained vesicles (Lee et al. 2011, Chivet et al. 2012, Rufino-Ramos et al. 2017), have been implicated as important vessels for transportation of cargo proteins between the CNS and periphery (reviewed recently in (Matsumoto et al. 2017a)), and recent work suggests this can also include key proteins involved with AD and PD such as tau (Shi et al. 2016a), Aβ42(Takahashi et al. 2002), and α-syn (Budnik et al. 2016, Coleman & Hill 2015, Thompson et al. 2016, Matsumoto et al. 2017b). Further, we have recently used immunoassay detection platforms to report that CNS-derived EVs within plasma contain altered levels of α-syn and tau in PD 95 (Shi et al. 2014). These studies hint at the potential utility of plasma EVs as a future biomarker resource that maintains the clinical ease of blood collection but avoids many of the complications involved with blood alone. However, isolation of CNS-derived EVs from plasma is a time-consuming and challenging process, and optimization is still needed in both EV isolation methods and assay development to assess the feasibility as a biomarker resource alternative.
Alternative platforms for protein biomarkers
Measurements of nearly all of the existing AD and PD molecular biomarker candidates identified in CSF and blood are carried out based on known pathogenically important proteins with antibody-based assays, which are often associated with relatively high variability, particularly when different detection techniques (different antibodies, sample preparation, calibrators, etc.) are used, leading to discrepant results across laboratories (Zetterberg 2015, Olsson et al. 2016, Chaloupka et al. 1996). Additionally, inter-laboratory standardization of immunoassay measurements of CSF tau and Aβ42 has proven difficult (Mattsson et al. 2011, Mattsson et al. 2013). Similar difficulties with immunoassay variability have been encountered in PD and related disorders (Kruse et al. 2015, Schapira 2013). Furthermore, development of quantitative immunoassays for most novel candidates identified by various exploratory approaches to look for protein candidates beyond known Aβ, tau and α-syn is limited by the lack of available (and specific) antibodies, which are further challenged by interference with antibody binding within complex solutions (e.g. biofluids such as CSF and blood) due to matrix effects. Additional obstacles are that de novo assay development is time consuming, prohibitively expensive for screening multiple markers, and very difficult to multiplex (Hoofnagle & Wener 2009). Thus, although a large library of potential protein biomarkers has been identified through proteomic studies (see below), the vast majority of candidates never reach the stage of validation (biomarker pipeline illustrated in Figure 1) and clinical testing (referred to as the “bottleneck” in biomarker development (Witkowska et al. 2012, Makawita & Diamandis 2010, Paulovich et al. 2008)). One approach to minimize the biomarker bottleneck could be use of alternate platforms, not limited by these challenges. As an antibody-free platform with high sensitivity, accuracy, and excellent multiplex capability, targeted mass spectrometry (MS) techniques such as selected reaction monitoring (SRM) may be an effective alternative for candidate validation. Or, alternatively as highlighted in this review, SRM can also work to reduce the bottle neck in biomarker development by providing a platform for efficient, and cost effective verification of large sets of candidates simultaneously.
Figure 1: Pipeline for biomarker development.
A pipeline for proteomics-based biomarker development typically proceeds through three preclinical stages: discovery, verification, and validation (Frantzi et al. 2014, Paulovich et al. 2008, Parker & Borchers 2014, Surinova et al. 2011, Rifai et al. 2006, Chan et al. 2016) with an inverse relationship between the numbers of subject samples and candidate markers investigated. Identification in the discovery phase is often determined by fold differences (i.e. up or down fold changes) in relative expression between disease and control samples within a few, often pooled samples. Then, candidates can enter the verification stage, where they can be further evaluated in a small set (10–50) of patient samples and critically analyzed for reproducibility and assay development. Once verified, a smaller panel of candidates (typically ≤ 10) can enter the validation stage, where they are validated within a much larger set (100–500) of samples (Parker & Borchers 2014) to assess the sensitivity and specificity as well as utility within a larger cohort. This is the final preclinical stage, and chosen candidates can be further evaluated for useful clinical application.
Mass Spectrometry
MS application to proteomics
In the last few decades, development and application of MS techniques have made essential contributions to proteomic studies involving protein identification (Altelaar et al. 2013), structure characterization (Konermann et al. 2011), as well as quantitation (Mallick et al. 2007, Peng et al. 2012, Angel et al. 2012). Until recently, MS has mostly been incorporated into large scale biomarker studies primarily through bottom-up, “shotgun” proteomic approaches. In these studies, proteins obtained from a sample are proteolytically cleaved into small peptides (classically by trypsin), which are fragmented within the MS to develop mass spectra that are then matched against a database, to ultimately deduce their protein of origin (Figure 2, see (Tsiatsiani & Heck 2015) for a descriptive review on MS technology). Peptide fragmentation patterns generated from tandem MS (often referred to as MS/MS, but will simply be referred to as MS in this review) add greatly to the reliability of protein/peptide identification and structure characterization. Coupling the MS system with liquid chromatography (LC) allows proteomic analysis of complex mixtures, as the linked LC system provides separation of proteins (and therefore improved resolution of MS) with significantly decreased sample purification and preparation time. MS-based proteomic analysis can also be quantitative, a fundamental necessity for identifying differences between biological samples in disease state characterization studies. Applications and limitations of the widely used quantitation strategies in LC-MS analysis have been reviewed in detail previously (see (Demmers 2012)). Briefly, peptides and proteins can be quantified by coupling isotope labels to amino acids by metabolic (Langen et al. 2000, Conrads et al. 2001, Ong et al. 2002, Beynon & Pratt 2005, Oda et al. 1999, Chahrour et al. 2015), chemical (Jin et al. 2016, Ross et al. 2004, Thompson et al. 2003), or enzymatic labeling (Heller et al. 2003, Yao et al. 2003, Mirgorodskaya et al. 2000, Bantscheff et al. 2004) strategies as well as more cost-effective label-free strategies (Neilson et al. 2011) such as ion intensity (Bondarenko et al. 2002, Ono et al. 2006) or spectrum counting analysis (Gilchrist et al. 2006, Liu et al. 2004, Otsuki et al. 2001) (See Figure 3A for quantitative MS approaches directly applicable to biomarker discovery workflows). Collectively, modern MS quantitation of proteins/peptides has been achieved in many proteomic studies, including neurodegenerative disorder related studies (Kitsou et al. 2008, Jin et al. 2006), which provided comparable, yet improved conclusions to those drawn from similar studies using conventional techniques (e.g., two-dimensional gel electrophoresis (2-DE)) (Basso et al. 2004), while also achieving greater depth of informative proteome analysis (Han et al. 2008).
Figure 2: Biomarker identification by top-down “shotgun” proteomics.
A: Typical process for extracting peptides from proteins within complex biological mixtures. After extracting proteins from biological sources, they are often separated (e.g. electrophoresis or column fractionation methods) before cleavage of proteins into peptide components by enzymatic digestion (most commonly by trypsin). B: Peptide identification by LC-MS. Following digestion, peptides are then further separated by liquid chromatography, ionized, and detected by the MS. Peptides are then further selected for fragmentation within the MS, and the individual components result in a fragmentation spectra identified by the detector (MS2). C: Process of protein identification. Fragmentation spectra can then be searched against spectral libraries of known peptide spectra to determine their peptide identity. Further bioinformatics can be applied to assess the validity of the peptides and resulting proteins identified by the study.
Figure 3: Select methods to quantify relative expression of protein biomarker candidates.
A: Isobaric chemical labeling for exploratory profiling. Isobaric labeling allows improved detection of peptides with fold difference in expression between a disease and control in complex biological mixtures for exploratory profiling in the discovery phase. Samples from each condition, which are digested and labeled separately prior to mixing and LC-MS analysis, are identified by MS2 fragmentation of isobaric labels, and the relative, quantitative difference can be used to determine differentially expressed peptide targets. B: Label-free methods for targeted approaches. SRM and PRM allow peptide fragmentation for improved relative quantitation of targeted species in complex biological mixtures for targeted-based biomarker verification. Samples containing digested peptides are analyzed separately by triple quadrapole LC-MS separately, where precursor ions are selected in the first quadrapole (Q1), fragmented within the second quadrapole (Q2), and analyzed within the third (Q3). For PRM, all transitions are measured within a HRAM Q-Orbitrap, while for SRM, specific combinations of the precursor ion with a product ion fragment (“transitions”) are selected for detection within the third quadrapole.
Advantages of MS as a platform for biomarker workflows
In addition to the high sensitivity and minimal analytical variation, protein identification by MS provides several technical advantages for protein biomarker workflows. First, it allows high-throughput generation of in-depth information on protein/peptide detection, even within complex mixtures like biological fluids. Second, peptides detected can include modified forms such as truncations and PTMs, which are thought to be important for AD, PD, and related diseases(Oueslati et al. 2010, Schmid et al. 2013, Barthelemy et al. 2016, Inoue et al. 2014, Vosseller 2007), and often require specialized and separate detection methods when using other platforms. Third, peptide detection is not reliant on affinity binding or antibody recognition, and therefore is both free of many analytical limitations by immunoaffinity based assays (e.g. ELISA), and can be additionally used in an unbiased approach for screening. Finally, further advantages of MS include its ability to distinguish between very closely related protein species, its ability to allow precise monitoring of biological and analytical variability due to sample handling, and its high multiplexing capacity (Hale 2013). Altogether, MS provides a powerful and highly sensitive peptide detection platform that is free of many analytical variations associated with other common antibody-based platforms.
Targeted MS technologies
Proteomic profiling techniques, while excellent for identifying potential biomarker candidates, are subject to their own difficulties in biomarker development (Frantzi et al. 2014), specifically that the widely used “shotgun” profiling techniques result in large pools of candidates including false discoveries (which may include false positive MS identifications and/or differences only detected in test cohorts) (Levitsky et al. 2017, Gupta & Pevzner 2009) so that each candidate requires further validation using independent methods. To meet this demand, modern MS techniques that allow selection and precise quantification of unique peptides, e.g., accurate inclusion mass screening (AIMS) (Jaffe et al. 2008) and selected reaction monitoring (SRM) (Whiteaker et al. 2011, Addona et al. 2009, Selevsek et al. 2011), have been developed. These targeted proteomic technologies have been proposed as the basis of a viable biomarker pipeline (Whiteaker et al. 2011) (Figure 1) and have become powerful tools in biomarker discovery due to their high sensitivity, accuracy and specificity. SRM, in particular, has emerged as an alternative to immunoaffinity-based measurements of defined protein sets with excellent reproducibility across different laboratories and instrument platforms (Addona et al. 2009, Selevsek et al. 2011).
SRM and PRM targeted MS approaches
SRM and parallel reaction monitoring (PRM) are two targeted MS approaches with powerful applications for biomarker detection and quantification (Figure 3B). In SRM analysis, precursor ions of different charge states are chosen as representative of their respective peptides for further investigation. Targeted precursor ions are initially filtered by the first quadrupole and then fragmented in the collision cell (second quadrupole). Then, in the third quadrupole, only specific product ions are selected for detection. Specific combinations of a precursor ion with a product ion are referred to as “SRM transitions”, which are transformed into a detectable signal for targeted peptide identification and quantification. Because fragmentation occurs within milliseconds, the collection window of a transition can be manipulated for each target (often referred to as “scheduled-SRM”) to increase the total number of targets added within a single run. Then, through careful extrapolation of the parent ions, the scheduled SRM approach can be applied for the identification and quantitation of hundreds of peptides in a single MS run (Kiyonami et al. 2011) (see (Calvo et al. 2011, Kuzyk et al. 2013, Boja & Rodriguez 2012, Gallien et al. 2011, Picotti & Aebersold 2012, Meng & Veenstra 2011, Vidova & Spacil 2017) for reviews of SRM technology and assay designs).
The PRM method performs comparably to SRM, but requires much less assay development (Ronsein et al. 2015). Using a high resolution accurate mass (HRAM) Q-Orbitrap MS, PRM filters and fragments precursor ions similarly to SRM but uses an Orbitrap mass analyzer to detect all resulting product ions (Wildsmith et al. 2014, Gallien et al. 2012, Bourmaud et al. 2016). As PRM has the advantages of acquiring full MS/MS spectrum of all fragment ions generated from the targeted peptides/proteins with high resolution and high mass accuracy, this approach has recently emerged as an alternative method of targeted quantitation (Rauniyar 2015, Peterson et al. 2012). This is especially useful for confirming the identity of the targeted peptides and characterizing associated PTMs. However, despite the improved resolution, PRM has a longer scan time for each targeted precursor compared to SRM, and therefore it is less efficient for targeting large sets of peptides simultaneously. Thus, to date, SRM remains the most common targeted MS approach for biomarker studies, and likely the most practical for large-scale candidate investigations.
To aid the targeted-MS assay development process, SRM is commonly supplemented with label-based quantitative approaches. This method provides the advantage of adding “heavy” isotope labeled internal standards (e.g. total 13C/15N labeled protein or specific peptides), which are chemically equivalent to the “light” endogenous counterparts and provide identical yield post preparation procedure, allowing easier identification of target peptides in complex mixtures (including those of low abundance or otherwise technically difficult to detect). These standards also function to further minimize analytical variation, as they maintain similar ionization and fragmentation behavior in the mass spectrometer, altogether minimizing impact on quantification errors due to variations in sample preparation and LC performance (Pannee et al. 2013). Finally, “spike-in” of heavy labeled standards also provides the ability to quantify (relative or absolute) endogenous peptides based on the light/heavy ratio within samples and directly compare relative expression across samples. All of these applications provide strong advantages for biomarker studies which utilize the SRM platform.
Application of SRM in biomarker studies
Altogether, targeted MS approaches like SRM convey the principle advantages of carrying out identification and quantitation of hundreds of protein targets, possess the capability of substantial multiplexing in a single run to increase the throughput, and, most importantly, maintain the ability to provide structural specificity, (Bereman et al. 2012) characteristics that make it valuable for biomedical research. Moreover, SRM allows measurement of relative and absolute peptide levels through detection of well-defined targeted peptides with high selectivity and sensitivity. Additionally, compared to routine immunoassays, SRM assays are highly flexible, more quickly developed, cost effective, and capable of distinguishing highly similar proteoforms such as PTMs (Vidova & Spacil 2017). And, like immunoassays, standard reference approaches have recently been developed and recognized for selective, accurate, and reproducible measurements of Aβ42 in CSF (Leinenbach et al. 2014, Korecka et al. 2014), comparable to the development and performance of AlzBio3 kit for immunoassay measurements (with potentially improved variability (Mattsson et al. 2009, Kuhlmann et al. 2017). Together, these advantages suggest SRM may be a suitable and cost effective platform for large scale candidate testing within the verification stage of biomarker development, as some studies have already demonstrated.
Application of SRM in AD biomarker studies
Application of SRM to verify previously reported candidate biomarkers is relatively new, but some studies have begun to utilize this technology in AD (Table 1). Commonly, workflows begin by identifying a list of potential targets based on previous reports demonstrating differential expression between disease and control, followed by assay development utilizing heavy isotope labeled standards to generate a panel of peptide targets for SRM targeting (see (Spellman et al. 2015) for an extensive collaborative development of multiplexed SRM peptide assays prior to follow up studies using heavy-labeled peptide standards). For example, Chen et al. applied SRM quantification along with stable isotope-labeled heavy protein internal standards to AD and control brain tissue samples and determined differences in concentrations of previously reported AD-relevant proteins, both between groups (Chen et al. 2013, Chen et al. 2012) and between sub-regions of the brain (Chen et al. 2012). Another group also looked across different brain regions in AD vs control tissue in an attempt to validate a panel of previously reported synaptic proteins, and reported differences in relative abundances between hippocampus and motor cortex for both AD and control (Chang et al. 2014). Similar to the tissue studies, other groups have begun using SRM aided by heavy labeled standards to verify candidate AD biomarkers within biofluids such as CSF. For example, Paterson et al. used SRM with heavy labeled peptides to assess a panel of 54 CSF biomarkers in two independent cohorts (which they grouped into ‘AD’ and ‘non-AD’ based on immunoassay measurement of total tau and Aβ42), and determined five proteins to be significantly elevated in AD in both cohorts (Paterson et al. 2016). A similar approach was taken in another recent study by Begcevic et al. attempting to verify a panel of 30 peptides in CSF of MCI, and AD patients further separated by AD dementia severity based on Mini-Mental State Examination (MMSE) and clinical dementia rating (CDR) scores, which reported several candidates that may correlate with AD severity, and possibly also disease progression (Begcevic et al. 2018). Another study by Wildsmith et al. quantified a panel of 39 peptides (from 30 proteins) within the CSF of control, MCI, and AD patients, and determined four targets were significantly different between AD and control, while four markers also demonstrated significant longitudinal change within AD (Wildsmith et al. 2014). Together, these studies provide examples of the practical application of SRM to verify panels of AD biomarker candidates. Each study began with a larger collection of previously reported candidates and utilized SRM to refine the candidate panels and highlight those which demonstrated practical biomarker applications.
Table 1:
Biomarker studies using SRM on human AD clinical samples.
| Source | MS Method | Purpose of MS in study | Finding by MS | Cohort | Reference |
|---|---|---|---|---|---|
| Brain tissue: Frontal and temporal cortex | SRM + heavy labeled quantification concatemer (QconCAT) | Quantification of clusterin in control vs severe AD. | Level of clusterin was significantly increased in AD vs control in frontal but not temporal cortex. | Frontal: 6 AD, 6 control Temporal: 6 AD, 6 control |
Chen et al. 2012 |
| Brain tissue: Frontal cortex | SRM+ heavy labeled quantification concatemer (QconCAT) | Quantification of total APP, APP695, and Aβ in control vs severe AD. | Level of Aβ was significantly increased in the AD vs control. Level of total APP was similar in AD vs control while APP695 was significantly lower AD vs control. | 10 AD, 7 control | Chen et al. 2013 |
| Brain tissue: Temporal cortex | SRM + internal heavy labeled peptide standards | Demonstrate the utility of SRM compared to other platforms, while also demonstrate histone acetylation changes within AD. | Histone acetylation significantly decreased in AD temporal lobe vs control. | 10 AD, 3 control | Zhang et al. 2012 |
| Brain tissue: Hippocampus and motor cortex | SRM + enolase as internal standard | Quantitative analysis of synaptic proteins in control vs AD. | Relative abundance of different subtypes of synaptic proteins in hippocampus vs motor cortex for both AD and control. | 6 AD, 6 control | Chang et al 2014 |
| CSF | SPE + SRM + heavy isotope labeled peptides | Absolute quantification of Aβ38, Aβ40, and Aβ42 in control vs AD. | Ratio of Aβ42/Aβ40 increased the separation performance between AD vs control as compared to Aβ42 alone. | Two sets of 15 AD vs 15 control | Pannee et al. 2013 |
| CSF | SRM + heavy stable isotope labeled peptides | Identification of longitudinally dynamic biomarkers in AD. | 4 CSF proteins were significantly different in AD vs control, while 4 demonstrated significant longitudinal changes in AD. | 45 AD, 5 MCI, 10 control | Wildsmith et al. 2014 |
| CSF | dMS + SRM | Discovery of candidate markers for AD progression. | Neurosecretory protein VGF and neuronal pentraxin receptor 1 were both significantly lower in AD vs controls. | Discovery cohort: 10 AD, 10 control Longitudinal cohort: 30 AD, 30 control |
Hendrickson et al. 2015 |
| CSF | SRM + heavy isotope labeled peptides | Validation of candidate biomarkers for AD progression in MCI vs AD (mild, moderate, and severe). | Neuronal pentraxin receptor 1 was significantly lower in AD vs MCI, as well as progressively lower in severe AD. | Set 1: 8 MCI, 11 Mild AD, 24 Moderate AD, 15 Severe AD Set 2: 6 MCI, 8 Mild AD, 16 Moderate AD, 13 Severe AD |
Begcevic et al. 2018 |
| CSF | SRM + high pH fractionation | Parallel Tau and Aβ42 detection by SRM. | Significant negative correlation between ratio of tau and Aβ peptides with disease severity. | 10 AD, 10 control, 10 MCI | Pottiez et al. 2017 |
| CSF | SRM + heavy isotope labelled peptide or yeast enolase peptide | Validation of candidate AD biomarkers. | 9 markers significantly elevated in AD patients vs control. | Cohort 1: 35 AD, 31 control. Cohort 2: 46 AD, 36 control |
Paterson et al. 2016 |
| CSF | SRM | Validation of a large peptide panel to discriminate AD-disease states. | 320 of 567 peptides in the MRM panel were detectable in > 10% of ADNI cohort used. | 66AD, 134 MCI, 85 control | Spellman et al. 2015 |
| Blood serum | SRM | Validation of serum ApoE protein levels in relation to AD. | ApoE level was lower in AD vs control. | 45 AD, 43 control | Han et al. 2014 |
| Tear fluid | LC-MS/MS profiling + SRM + heavy isotope labeled peptides | Discovery and verification of AD biomarkers within tear fluids. | 6 markers significantly altered in AD vs control, of which a panel of four targets was reported with greatest potential diagnostic utility. | 14 AD, 9 control | Kalló et al. 2016 |
Aβ = amyloid beta, AD = Alzheimer’s disease, ApoE = Apolipoprotein E, APP = amyloid precursor protein, CSF = cerebral spinal fluid, dMS = differential MS, LC-MS/MS = liquid chromatography tandem mass spectrometry, MCI = mild cognitive impairment, SPE = solid phase extraction, SRM = selected reaction monitoring.
In addition to marker verification, some studies have also used SRM coupled with heavy labeled standards to examine current AD biomarker peptides closely and determine their absolute quantification within groups. Pannee et al. determined the absolute quantification of Aβ peptides (Aβ38, Aβ40, Aβ42) in the CSF of AD and control patients by coupling solid-phase peptide extraction to SRM with heavy labeled peptide standards, and reported similar disease-association efficacy of SRM identified Aβ42 to that of traditional ELISA assays (which was further enhanced when taking the Aβ42/ Aβ40 ratio) (Pannee et al. 2013). A study by Han et al. validated serum ApoE protein levels in relation to AD using SRM MS-based analysis from the blood serum of control and AD patients, and further compared these results to a parallel measure using fluorescence-activated cell sorting (FACS) analysis, and reported comparably decreased levels in AD vs control (Han et al. 2014). Finally, we applied a high pH fractionation peptide separation method (high-pressure, high-resolution separations coupled with intelligent selection and multiplexing, a.k.a. PRISM) (Shi et al. 2012) to an SRM with heavy peptide standard workflow to determine the absolute CSF concentrations of tau and Aβ42 in parallel within control, MCI, and AD groups, and demonstrated their efficacy to separate groups. We found equivalent diagnostic utility to immunoassay measurements within the same sample set, with particular strength in the ratio of Aβ42 to global Aβ expression (Pottiez et al. 2017). Together, these studies illustrate the capacity of SRM to apply both relative and absolute quantification of peptide targets for verification of AD candidate biomarkers.
Applications of SRM for PD biomarker verification
To a lesser extent than AD, some studies have also attempted to verify biomarker candidates for PD (Table 2), using workflows similar to those described above. Building off previous proteomic profiling, bioinformatics analysis, and SRM assay optimization, we developed a refined panel of protein peptide targets differentially expressed between disease and control which were further verified by SRM to provide useful marker panels for PD diagnosis (5 peptide panel) and disease severity correlation (2 peptide panel)(Shi et al. 2015), however larger cohort and longitudinal validation studies are needed. In a separate study, we also developed a list of potential glycopeptide candidate targets based on previous reports of differential expression within brain tissue and CSF, and developed assays for SRM targeting within plasma samples of PD and control (Pan et al. 2014, Hwang et al. 2010). Along with demonstrating the practicality of SRM for detecting low abundance biomarkers in the periphery with the aid of heavy labeled standard peptides, differential comparison of PD and healthy controls, along with Unified Parkinson’s Disease Rating Scale (UPDRS) scores, revealed potential verification of glycosylation PTMs as markers of PD progression, although further studies in larger, independent cohorts are needed for more complete interpretation (independent cohort validation of both studies is currently ongoing in our lab) (Pan et al. 2014). In another study, after previously reporting a panel of 13 proteins extracted from T-lymphocytes and identified by 2-DE as a discriminant signature for PD (Alberio et al. 2012), control, or atypical parkinsonism, Alberio et al. also used SRM to verify this molecular signature in an independent cohort and platform. Using 9 selected peptides (from 7 proteins), 8 displayed a fold change in agreement with 2-DE. Additionally, using a discriminant classification system to adequately compare the two methods (with respect to modified and total protein detection), 16 of the 18 patients were properly classified (Alberio et al. 2014). Although the sample size was low for verification as we and others have described (Rifai et al. 2006, Surinova et al. 2011, Parker & Borchers 2014), this study particularly highlights the additional practicality of utilizing SRM to verify biomarker candidates discovered by multiple platforms.
Table 2:
Biomarker studies using SRM on human PD clinical samples.
| Source | MS Method | Purpose of MS in study | Finding by MS | Cohort | Reference |
|---|---|---|---|---|---|
| CSF | 2D-LC-MS/MS + iTRAQ + SRM | 2D-LC-MS/MS + iTRAQ: Identify biomarker candidates differentially regulated between PD, PDD, and control. SRM: Verify identified candidates. |
16 differentially regulated proteins between PD and PDD by ITRAQ, only 2 PD vs control or PDD vs control could be verified by SRM. | 12 PD, 12 PDD, and 12 control (pooled) | Lehnert et al. 2012 |
| Plasma | SRM + heavy isotope labeled peptides |
Discover and verify panel of glyocopeptides which may correlate with PD diagnosis or severity. | 3 glycopeptides differentially expressed between diagnostic groups; panel of four correlated with diagnosis, panel of 2 with PD severity. | Initial validation: 15 PD, 15 AD, 15 control (pooled from 75 PD, 15 AD, 30 control) Final validation: 98 PD, 15 AD, 49 control |
Pan et al. 2014 |
| Blood | SRM + single (pre-digested) standard spike in | Verify PD-protein signature in T-lymphocytes identified by 2-DE with SRM in an independent cohort. | 8 of 9 selected peptides displayed fold change concordant with 2-DE discovery study. | 9 PD vs 9 control | Alberio et al. 2014 |
| CSF | 2D-LC-MS/MS + SRM + heavy peptide spike in | Identify and verify peptides which could correlate with PD diagnosis or severity. | ~1400 peptides differentially expressed in PD vs normal in training set, Five peptide panel able to differentiate PD vs control or AD, two which correlated with PD severity. |
Training: 30 PD vs 30 control (pooled) Validation: 40 PD vs 38 AD vs 40 control |
Shi et al. 2015 |
| CSF | SRM + fractionation + heavy protein spike in of α-syn | Measure absolute α-syn and validate immunoassay reported differences among PD vs related diseases. | αSyn, βSyn and γSyn concentrations were increased in AD and Creutzfeldt-Jakob disease but not altered in PD, PDD, LBD and atypical parkinsonian syndromes. | 37 Control, 23 PD, 17 PDD, 10 LBD, 20 PSP, 10 CBS, 19 AD, 10 CJD | Oeckl et al. 2016 |
| CSF | SRM + heavy protein spike in of α-syn | Measure absolute α-syn in CSF and identify novel peptide markers. | Six peptides of α-syn identified, one novel peptide correlated with PD severity, and changes significantly tracked progression longitudinally. | Cross sectional: 15 PD vs 15 control Longitudinal: 15 PD for 2 points |
Yang et al. 2017 |
2-DE = two-dimensional gel electrophoresis α-syn = alpha-synuclein, AD: Alzheimer´s disease, CBS: corticobasal syndrome, CJD: Creutzfeldt-Jakob disease, CSF = cerebral spinal fluid, LBD: Lewy body dementia, LC-MS/MS = liquid chromatography tandem mass spectrometry, PD: Parkinson´s disease, PDD: Parkinson’s disease with dementia, PSP: progressive supranuclear palsy, SRM = selected reaction monitoring.
As with Aβ42 and tau for AD studies, SRM has also been utilized to closely monitor the absolute quantification of α-syn in relation to PD studies. Using a peptide fractionation method followed by SRM targeting aided by heavy standards, Oeckl. et al. measured α-syn primarily in its unmodified form (spanning roughly 70% of the protein) within various neurodegenerative disease groups, and found no alterations in α-syn expression within PD or PD related diseases compared to control, a result that contradicts most immunoassay studies, possibly resulting from the lack of C-terminal and/ or modified peptides included in the study (Oeckl et al. 2016). We similarly reported an SRM assay for measuring absolute α-syn quantification without fractionation (Yang et al. 2017), and found one peptide was particularly strong in differentiating PD from control with similar power to immunoassay detection (Hong et al. 2010, Wang et al. 2012). Like the Aβ peptide study by Panne et al. and our study with parallel Aβ and tau detection, these studies demonstrate the potential utility of absolute quantification by SRM to closely monitor biomarker peptides with both well-established roles in pathological development as well as prior studies using traditional detection platforms for comparative assessment. Further studies are still needed to confirm the validity of these peptide targets as accurate reporters of total protein presence. Additionally, more studies measuring these targets by both traditional and SRM platforms in parallel are needed to accurately assess the advantages of utilizing this approach moving forward. Further, the potential to develop assays that specifically target proteoforms relevant for protein aggregation and or pathological development, such as aggregated or modified α-syn (e.g. phosphorylated serine 129), is a strong advantage for SRM over immunoassay development, perhaps providing a platform for parallel detection of multiple unmodified, modified, and aggregated targets of a marker without the limitations associated with antibody capture.
Bridging biomarker discovery and verification with SRM
Another advantage of MS approaches in the biomarker pipeline is that platforms for discovery and verification can be coupled together within a single study (see Figure 4 for an illustrated workflow). Indeed, some studies have worked to combine the strengths of deep exploratory profiling, quantitative labeling, and targeted-MS approaches to both discover and further verify biomarkers for AD and PD. For example, after applying iTRAQ isobaric labeling to CSF samples from healthy controls and PD patients with or without dementia, Lehnert et al. first used shotgun proteomics in a discovery phase to determine differentially expressed peptides for candidate targets, then applied targeted SRM proteomic analysis in a separate verification phase within a single study. After identifying a panel of differentially expressed proteins between groups in the discovery study, the panel was fine-tuned to only a few candidates following SRM verification (Lehnert et al. 2012). As mentioned above, a study in our lab presented a staged pipeline for biomarker development taking a similar approach, coupling unbiased data-dependent acquisition (DDA) analysis of MS with the high sensitivity, accuracy and specificity of SRM analysis to identify and subsequently verify biomarkers from CSF samples of PD patients compared to healthy controls (Shi et al. 2015). Similarly to Lehnert et al., we discovered a list of candidates that were substantially altered in PD subjects, which was further fine-tuned following SRM verification within the single study. In a slightly different approach, Hendrickson et al applied a high resolution open discovery platform (coined ‘differential MS’), which, rather than isobaric labeling, takes into account the variability of measured intensities in both high and low abundance ions (Wiener et al. 2004) and allows relative quantification without an internal standard, first within a small discovery cohort of AD and control to identify differentially expressed candidate markers. Again, in the same study, two markers were further selected for a SRM targeting in a larger, longitudinal cohort, and the authors reported an estimated annual rate of differential expression per year in AD (Hendrickson et al. 2015). Altogether, these studies demonstrate the practical application of coupled discovery and verification through MS platforms, and provide a path for future biomarker studies moving forward. Although advancements in discovery platforms (including MS) has greatly enhanced sensitivity of candidate identification, these and other studies highlight the critical need for systematic workflows that subsequently challenge and refine such candidates prior to moving onto costly stages of validation testing. Targeted MS approaches like SRM provide such a platform, which can be efficiently utilized to verify previously reported panels of targets, or ideally, coupled within a discovery study to refine the final panel reported.
Figure 4: Proposed workflow of coupled unbiased discovery and targeted verification of biomarkers by MS within a single study.
In the first discovery phase (Phase 1), a small set of samples are isobarically labeled and processed for deep exploratory profiling to quantitatively identify peptides which display increased or decreased fold differences in a disease vs control setting. Then, in a follow-up verification phase (Phase 2) in a larger cohort, a panel of candidates from the discovery phase is rigorously tested for verification by a targeted SRM approach supplemented by “spike-in” of heavy standard peptides for both detection aid and relative quantification.
Future perspective
Utilizing SRM to improve AD and PD biomarker specificity
Previous efforts have identified panels of promising markers for improved diagnostic separation of AD or PD from their respective controls. As diagnosis is currently heavily dependent on symptomatic presentation, confirmation of diagnosis at autopsy demonstrates a high rate of misdiagnosis based on clinical symptoms 8,172–178, particularly at early disease stages (Tolosa et al. 2006, Dubois et al. 2016). Other forms of dementia such as frontotemporal dementia (FTD) and those associated with PD and cerebrovascular disease, are often confused with AD, and sufficient evidence to confirm the diagnosis is lacking, as 12–23% of patients classified as AD during life are not confirmed at autopsy (Gaugler et al. 2013, Klatka et al. 1996, Pearl 1997, Ranginwala et al. 2008, Lim et al. 1999). Similarly, other parkinsonian diseases and motor disorders are often falsely diagnosed as PD, with up to 25% of PD diagnosed patients alternatively diagnosed post-mortem (Tolosa et al. 2006, Hughes et al. 1992, Marsili et al. 2018). Therefore, in addition to developing improved markers for early detection and tracking of progression, improvements are also needed to increase their specificity to decrease the chance of misdiagnosis (Dubois et al. 2016). The utility of currently available molecular biomarkers (e.g., Aβ42 and tau for AD and α-syn for PD) in such differential diagnosis has shown to be limited, probably due to overlap in the underlying primary pathologies, and introduction of additional biomarkers reflecting other types of pathologies could be of value to optimize the differential diagnosis. With its antibody free, highly multiplex capacity, SRM provides a powerful platform for the simultaneous detection of panels of markers, which, when measured together, may improve differentiation between AD and other dementias or PD and related disorders (an approach currently underway in our laboratory).
SRM to improve pathological biomarkers in the blood through EVs
As discussed prior, the most promising biomarkers for both AD and PD are measured within CSF, an impractical resource for pre-clinical or prodromal patients within a clinical setting. Blood, which is a practical alternative, has its own challenges (reviewed in (Thambisetty & Lovestone 2010)), as extensive studies have indicated the potential challenges of diagnostic or predictive blood-based biomarkers for AD and PD due to extraordinarily low-abundance of disease-specific proteins able to be detected (Hong et al. 2010, Lin et al. 2012, Snyder et al. 2014, Henriksen et al. 2014, Rembach et al. 2014, Swaminathan et al. 2014, Lovheim et al. 2017). Thus, transitioning to blood remains challenging and likely dependent on technological advances. However, as also mentioned above, isolation of blood based but CNS-derived EVs may be a more efficient application of blood as a biomarker resource. Along with allowing multiple markers to be measured in a single preparation, another major advantage SRM could provide is the detection of PTM modified cargo such as α-syn thought to be highly abundant within EVs (reviewed in (Moreno-Gonzalo et al. 2014)). As proteins like α-syn and tau are thought to be highly modified by PTMs during aggregation (Jho et al. 2010, Morris et al. 2011, Andringa et al. 2004, Paleologou et al. 2010, Lashuel et al. 2013), SRM may provide a powerful platform for the parallel and simultaneous detection of multiple pathological forms (PTM, truncated, aggregated, etc.) originating from the CNS but accessible within the blood. Although sample isolation and assay developments still require optimization (currently ongoing in our lab), this SRM application has the capacity to utilize the blood as a resource for detecting pathological progression well beyond the capability of immunoassays.
Conclusions
Improvements are needed in both the sensitivity and specificity of current biomarkers for AD and PD. Application of exploratory MS-techniques has increased the quantity of reported candidate biomarkers for AD, PD and many other diseases; however, a notable distinction between the discovery and validation phases of biomarker development still remains. Much of the criticism in the biomarker field results from the discrepancy between discovery and validation, in which a validated marker has a defined clinical utility demonstrated across multiple patient cohorts (Crutchfield et al. 2016). Both phases are challenging and expensive, and validation requires an extra depth of resources (Horvatovich & Bischoff 2010). Targeted MS methods like SRM may provide a platform for large-scale candidate verification prior to costly validation testing (such as the studies highlighted in Tables 1 and 2). Further, we propose the use of parallel exploratory and targeted-MS approaches within a single study as a method for improving the bottleneck to biomarker validation within the AD and PD biomarker fields.
Acknowledgements and conflict of interest disclosure
The authors were supported by grants from the National Institutes of Health (NIH) (U01 NS082137, U01 NS091272, R01 AG056711, and R56 AG057417 to J.Z). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.Nothing to disclose.
Abbreviations used
- 2-DE
Two-dimensional gel electrophoresis
- α-syn
Alpha-synuclein
- Aβ
Amyloid beta
- AD
Alzheimer’s disease
- CNS
Central Nervous System
- CSF
Cerebral spinal fluid
- DA
Dopaminergic
- DLB
Dementia with Lewy body
- ELISA
Enzyme-linked immunosorbent assay
- EV
Extracellular vesicle
- FTD
Frontotemporal dementia
- LC-MS/MS
Liquid chromatography tandem mass spectrometry
- ND
Neurodegenerative diseases
- MCI
Mild cognitive impairment
- MS
Mass spectrometry
- MRI
Magnetic resonance imaging
- MRM
Multiple reaction monitoring
- PD
Parkinson’s disease
- PET
Positron imaging tomography
- PRISM
High-pressure, high-resolution separations with intelligent selection and multiplexing
- PRM
Parallel reaction monitoring
- PTM
Post-translational modification
- SN
Substantia nigra
- SRM
Selected reaction monitoring
References
- Abdo WF, Bloem BR, Van Geel WJ, Esselink RA and Verbeek MM (2007) CSF neurofilament light chain and tau differentiate multiple system atrophy from Parkinson’s disease. Neurobiol Aging, 28, 742–747. [DOI] [PubMed] [Google Scholar]
- Addona TA, Abbatiello SE, Schilling B et al. (2009) Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma. Nat. Biotechnol, 27, 633–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alberio T, McMahon K, Cuccurullo M, Gethings LA, Lawless C, Zibetti M, Lopiano L, Vissers JP and Fasano M (2014) Verification of a Parkinson’s disease protein signature in T-lymphocytes by multiple reaction monitoring. Journal of proteome research, 13, 3554–3561. [DOI] [PubMed] [Google Scholar]
- Alberio T, Pippione AC, Zibetti M, Olgiati S, Cecconi D, Comi C, Lopiano L and Fasano M (2012) Discovery and verification of panels of T-lymphocyte proteins as biomarkers of Parkinson’s disease. Scientific reports, 2, 953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alonso A, Zaidi T, Novak M, Grundke-Iqbal I and Iqbal K (2001) Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments. Proc. Natl. Acad. Sci. U. S. A, 98, 6923–6928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alonso AC, Grundke-Iqbal I and Iqbal K (1996) Alzheimer’s disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules. Nat. Med, 2, 783–787. [DOI] [PubMed] [Google Scholar]
- Altelaar AF, Munoz J and Heck AJ (2013) Next-generation proteomics: towards an integrative view of proteome dynamics. Nat Rev Genet, 14, 35–48. [DOI] [PubMed] [Google Scholar]
- An C, Pu X, Xiao W and Zhang H (2018) Expression of the DJ-1 protein in the serum of Chinese patients with Parkinson’s disease. Neurosci Lett, 665, 236–239. [DOI] [PubMed] [Google Scholar]
- Anderson DC and Kodukula K (2014) Biomarkers in pharmacology and drug discovery. Biochemical pharmacology, 87, 172–188. [DOI] [PubMed] [Google Scholar]
- Andringa G, Lam KY, Chegary M, Wang X, Chase TN and Bennett MC (2004) Tissue transglutaminase catalyzes the formation of alpha-synuclein crosslinks in Parkinson’s disease. FASEB J, 18, 932–934. [DOI] [PubMed] [Google Scholar]
- Angel TE, Aryal UK, Hengel SM, Baker ES, Kelly RT, Robinson EW and Smith RD (2012) Mass spectrometry-based proteomics: existing capabilities and future directions. Chemical Society reviews, 41, 3912–3928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D and Jones E (2011) Alzheimer’s disease. Lancet, 377, 1019–1031. [DOI] [PubMed] [Google Scholar]
- Bantscheff M, Dumpelfeld B and Kuster B (2004) Femtomol sensitivity post-digest (18)O labeling for relative quantification of differential protein complex composition. Rapid Commun Mass Spectrom, 18, 869–876. [DOI] [PubMed] [Google Scholar]
- Barrett PJ and Timothy Greenamyre J (2015) Post-translational modification of alpha-synuclein in Parkinson’s disease. Brain Res, 1628, 247–253. [DOI] [PubMed] [Google Scholar]
- Barthelemy NR, Gabelle A, Hirtz C et al. (2016) Differential Mass Spectrometry Profiles of Tau Protein in the Cerebrospinal Fluid of Patients with Alzheimer’s Disease, Progressive Supranuclear Palsy, and Dementia with Lewy Bodies. J Alzheimers Dis, 51, 1033–1043. [DOI] [PubMed] [Google Scholar]
- Basso M, Giraudo S, Corpillo D, Bergamasco B, Lopiano L and Fasano M (2004) Proteome analysis of human substantia nigra in Parkinson’s disease. Proteomics, 4, 3943–3952. [DOI] [PubMed] [Google Scholar]
- Basurto-Islas G, Luna-Munoz J, Guillozet-Bongaarts AL, Binder LI, Mena R and Garcia-Sierra F (2008) Accumulation of aspartic acid421- and glutamic acid391-cleaved tau in neurofibrillary tangles correlates with progression in Alzheimer disease. J. Neuropathol. Exp. Neurol, 67, 470–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beach TG, Schneider JA, Sue LI, Serrano G, Dugger BN, Monsell SE and Kukull W (2014) Theoretical impact of Florbetapir (18F) amyloid imaging on diagnosis of alzheimer dementia and detection of preclinical cortical amyloid. J Neuropathol Exp Neurol, 73, 948–953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Begcevic I, Tsolaki M, Brinc D et al. (2018) Neuronal pentraxin receptor-1 is a new cerebrospinal fluid biomarker of Alzheimer’s disease progression. F1000Res, 7, 1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bereman MS, MacLean B, Tomazela DM, Liebler DC and MacCoss MJ (2012) The development of selected reaction monitoring methods for targeted proteomics via empirical refinement. Proteomics, 12, 1134–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beynon RJ and Pratt JM (2005) Metabolic labeling of proteins for proteomics. Mol Cell Proteomics, 4, 857–872. [DOI] [PubMed] [Google Scholar]
- Biomarkers Definitions Working, G. (2001) Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther, 69, 89–95. [DOI] [PubMed] [Google Scholar]
- Blennow K, Hampel H, Weiner M and Zetterberg H (2010) Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nature reviews. Neurology, 6, 131–144. [DOI] [PubMed] [Google Scholar]
- Blennow K and Zetterberg H (2015) The past and the future of Alzheimer’s disease CSF biomarkers-a journey toward validated biochemical tests covering the whole spectrum of molecular events. Front Neurosci, 9, 345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boccardi M, Altomare D, Ferrari C et al. (2016) Assessment of the Incremental Diagnostic Value of Florbetapir F 18 Imaging in Patients With Cognitive Impairment: The Incremental Diagnostic Value of Amyloid PET With [18F]-Florbetapir (INDIA-FBP) Study. JAMA Neurol, 73, 1417–1424. [DOI] [PubMed] [Google Scholar]
- Boja ES and Rodriguez H (2012) Mass spectrometry-based targeted quantitative proteomics: achieving sensitive and reproducible detection of proteins. Proteomics, 12, 1093–1110. [DOI] [PubMed] [Google Scholar]
- Bondarenko PV, Chelius D and Shaler TA (2002) Identification and relative quantitation of protein mixtures by enzymatic digestion followed by capillary reversed-phase liquid chromatography-tandem mass spectrometry. Anal Chem, 74, 4741–4749. [DOI] [PubMed] [Google Scholar]
- Bonifati V, Rizzu P, van Baren MJ et al. (2003) Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science, 299, 256–259. [DOI] [PubMed] [Google Scholar]
- Bourmaud A, Gallien S and Domon B (2016) Parallel reaction monitoring using quadrupole-Orbitrap mass spectrometer: Principle and applications. Proteomics, 16, 2146–2159. [DOI] [PubMed] [Google Scholar]
- Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN and Braak E (2003) Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging, 24, 197–211. [DOI] [PubMed] [Google Scholar]
- Breydo L, Wu JW and Uversky VN (2012) Alpha-synuclein misfolding and Parkinson’s disease. Biochim Biophys Acta, 1822, 261–285. [DOI] [PubMed] [Google Scholar]
- Broussolle E, Dentresangle C, Landais P et al. (1999) The relation of putamen and caudate nucleus 18F-Dopa uptake to motor and cognitive performances in Parkinson’s disease. J Neurol Sci, 166, 141–151. [DOI] [PubMed] [Google Scholar]
- Budnik V, Ruiz-Canada C and Wendler F (2016) Extracellular vesicles round off communication in the nervous system. Nat. Rev. Neurosci, 17, 160–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calvo E, Camafeita E, Fernandez-Gutierrez B and Lopez JA (2011) Applying selected reaction monitoring to targeted proteomics. Expert review of proteomics, 8, 165–173. [DOI] [PubMed] [Google Scholar]
- Chahrour O, Cobice D and Malone J (2015) Stable isotope labelling methods in mass spectrometry-based quantitative proteomics. J Pharm Biomed Anal, 113, 2–20. [DOI] [PubMed] [Google Scholar]
- Chaloupka JC, Putman CM, Citardi MJ, Ross DA and Sasaki CT (1996) Endovascular therapy for the carotid blowout syndrome in head and neck surgical patients: diagnostic and managerial considerations. AJNR Am J Neuroradiol, 17, 843–852. [PMC free article] [PubMed] [Google Scholar]
- Chan PP, Wasinger VC and Leong RW (2016) Current application of proteomics in biomarker discovery for inflammatory bowel disease. World J Gastrointest Pathophysiol, 7, 27–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang RY, Etheridge N, Dodd PR and Nouwens AS (2014) Targeted quantitative analysis of synaptic proteins in Alzheimer’s disease brain. Neurochem Int, 75, 66–75. [DOI] [PubMed] [Google Scholar]
- Chen J, Wang M and Turko IV (2012) Mass spectrometry quantification of clusterin in the human brain. Mol Neurodegener, 7, 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J, Wang M and Turko IV (2013) Quantification of amyloid precursor protein isoforms using quantification concatamer internal standard. Anal Chem, 85, 303–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng HC, Ulane CM and Burke RE (2010) Clinical progression in Parkinson disease and the neurobiology of axons. Annals of neurology, 67, 715–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chivet M, Hemming F, Pernet-Gallay K, Fraboulet S and Sadoul R (2012) Emerging role of neuronal exosomes in the central nervous system. Frontiers in physiology, 3, 145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coleman BM and Hill AF (2015) Extracellular vesicles--Their role in the packaging and spread of misfolded proteins associated with neurodegenerative diseases. Semin. Cell Dev. Biol, 40, 89–96. [DOI] [PubMed] [Google Scholar]
- Compta Y, Marti MJ, Ibarretxe-Bilbao N, Junque C, Valldeoriola F, Munoz E, Ezquerra M, Rios J and Tolosa E (2009) Cerebrospinal tau, phospho-tau, and beta-amyloid and neuropsychological functions in Parkinson’s disease. Mov Disord, 24, 2203–2210. [DOI] [PubMed] [Google Scholar]
- Conrads TP, Alving K, Veenstra TD et al. (2001) Quantitative analysis of bacterial and mammalian proteomes using a combination of cysteine affinity tags and 15N-metabolic labeling. Anal Chem, 73, 2132–2139. [DOI] [PubMed] [Google Scholar]
- Coon DW and Edgerly ES (1999) The personal and social consequences of Alzheimer disease. Genet Test, 3, 29–36. [DOI] [PubMed] [Google Scholar]
- Cooper AA, Gitler AD, Cashikar A et al. (2006) Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson’s models. Science, 313, 324–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costa OR, Verhaeghen K, Roels S, Stange G, Ling Z, Pipeleers D, Gorus FK and Martens GA (2018) An analytical comparison of three immunoassay platforms for subpicomolar detection of protein biomarker GAD65. PLoS One, 13, e0193670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crutchfield CA, Thomas SN, Sokoll LJ and Chan DW (2016) Advances in mass spectrometry-based clinical biomarker discovery. Clinical proteomics, 13, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cummings JL and Cole G (2002) Alzheimer disease. JAMA, 287, 2335–2338. [DOI] [PubMed] [Google Scholar]
- de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL and Hyman BT (2010) Caspase activation precedes and leads to tangles. Nature, 464, 1201–1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Demmers JAA S. KA (2012) Labeling Methods in Mass Spectrometry Based Quantitative Proteomics. (Leung DH-C ed.). [Google Scholar]
- Desikan RS, Cabral HJ, Settecase F et al. (2010) Automated MRI measures predict progression to Alzheimer’s disease. Neurobiol. Aging, 31, 1364–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dubois B, Feldman HH, Jacova C et al. (2007) Research criteria for the diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA criteria. Lancet Neurol, 6, 734–746. [DOI] [PubMed] [Google Scholar]
- Dubois B, Padovani A, Scheltens P, Rossi A and Dell’Agnello G (2016) Timely Diagnosis for Alzheimer’s Disease: A Literature Review on Benefits and Challenges. J Alzheimers Dis, 49, 617–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duran R, Barrero FJ, Morales B, Luna JD, Ramirez M and Vives F (2010) Plasma alpha-synuclein in patients with Parkinson’s disease with and without treatment. Mov Disord, 25, 489–493. [DOI] [PubMed] [Google Scholar]
- El-Agnaf OM, Salem SA, Paleologou KE, Curran MD, Gibson MJ, Court JA, Schlossmacher MG and Allsop D (2006) Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J, 20, 419–425. [DOI] [PubMed] [Google Scholar]
- Fagan AM, Roe CM, Xiong C, Mintun MA, Morris JC and Holtzman DM (2007) Cerebrospinal fluid tau/beta-amyloid(42) ratio as a prediction of cognitive decline in nondemented older adults. Arch Neurol, 64, 343–349. [DOI] [PubMed] [Google Scholar]
- Fairfoul G, McGuire LI, Pal S et al. (2016) Alpha-synuclein RT-QuIC in the CSF of patients with alpha-synucleinopathies. Ann Clin Transl Neurol, 3, 812–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferreon AC and Deniz AA (2007) Alpha-synuclein multistate folding thermodynamics: implications for protein misfolding and aggregation. Biochemistry, 46, 4499–4509. [DOI] [PubMed] [Google Scholar]
- Findley LJ (2007) The economic impact of Parkinson’s disease. Parkinsonism Relat Disord, 13 Suppl, S8–S12. [DOI] [PubMed] [Google Scholar]
- Forsberg A, Engler H, Almkvist O, Blomquist G, Hagman G, Wall A, Ringheim A, Langstrom B and Nordberg A (2008) PET imaging of amyloid deposition in patients with mild cognitive impairment. Neurobiol. Aging, 29, 1456–1465. [DOI] [PubMed] [Google Scholar]
- Frantzi M, Bhat A and Latosinska A (2014) Clinical proteomic biomarkers: relevant issues on study design & technical considerations in biomarker development. Clin Transl Med, 3, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gallien S, Duriez E, Crone C, Kellmann M, Moehring T and Domon B (2012) Targeted proteomic quantification on quadrupole-orbitrap mass spectrometer. Mol Cell Proteomics, 11, 1709–1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gallien S, Duriez E and Domon B (2011) Selected reaction monitoring applied to proteomics. Journal of mass spectrometry : JMS, 46, 298–312. [DOI] [PubMed] [Google Scholar]
- Gaugler JE, Ascher-Svanum H, Roth DL, Fafowora T, Siderowf A and Beach TG (2013) Characteristics of patients misdiagnosed with Alzheimer’s disease and their medication use: an analysis of the NACC-UDS database. BMC Geriatr, 13, 137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghidoni R, Paterlini A and Benussi L (2013) Translational proteomics in Alzheimer’s disease and related disorders. Clinical biochemistry, 46, 480–486. [DOI] [PubMed] [Google Scholar]
- Gilchrist A, Au CE, Hiding J et al. (2006) Quantitative proteomics analysis of the secretory pathway. Cell, 127, 1265–1281. [DOI] [PubMed] [Google Scholar]
- Giraldez-Perez R, Antolin-Vallespin M, Munoz M and Sanchez-Capelo A (2014) Models of alpha-synuclein aggregation in Parkinson’s disease. Acta Neuropathol Commun, 2, 176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goetzl EJ, Schwartz JB, Abner EL, Jicha GA and Kapogiannis D (2018) High complement levels in astrocyte-derived exosomes of Alzheimer’s disease. Annals of neurology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groveman BR, Orru CD, Hughson AG et al. (2018) Rapid and ultra-sensitive quantitation of disease-associated alpha-synuclein seeds in brain and cerebrospinal fluid by alphaSyn RT-QuIC. Acta Neuropathol Commun, 6, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gupta N and Pevzner PA (2009) False discovery rates of protein identifications: a strike against the two-peptide rule. J Proteome Res, 8, 4173–4181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hale JE (2013) Advantageous uses of mass spectrometry for the quantification of proteins. Int J Proteomics, 2013, 219452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall S, Ohrfelt A, Constantinescu R et al. (2012) Accuracy of a panel of 5 cerebrospinal fluid biomarkers in the differential diagnosis of patients with dementia and/or parkinsonian disorders. Arch Neurol, 69, 1445–1452. [DOI] [PubMed] [Google Scholar]
- Hall S, Surova Y, Ohrfelt A, Zetterberg H, Lindqvist D and Hansson O (2015) CSF biomarkers and clinical progression of Parkinson disease. Neurology, 84, 57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halliday GM, Holton JL, Revesz T and Dickson DW (2011) Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol, 122, 187–204. [DOI] [PubMed] [Google Scholar]
- Hampel H, Blennow K, Shaw LM, Hoessler YC, Zetterberg H and Trojanowski JQ (2010) Total and phosphorylated tau protein as biological markers of Alzheimer’s disease. Experimental gerontology, 45, 30–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hampel H, Burger K, Teipel SJ, Bokde AL, Zetterberg H and Blennow K (2008) Core candidate neurochemical and imaging biomarkers of Alzheimer’s disease. Alzheimer’s & dementia : the journal of the Alzheimer’s Association, 4, 38–48. [DOI] [PubMed] [Google Scholar]
- Hampel H, Lista S and Khachaturian ZS (2012) Development of biomarkers to chart all Alzheimer’s disease stages: the royal road to cutting the therapeutic Gordian Knot. Alzheimers Dement, 8, 312–336. [DOI] [PubMed] [Google Scholar]
- Han SH, Kim JS, Lee Y et al. (2014) Both targeted mass spectrometry and flow sorting analysis methods detected the decreased serum apolipoprotein E level in Alzheimer’s disease patients. Mol Cell Proteomics, 13, 407–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han X, Aslanian A and Yates JR 3rd (2008) Mass spectrometry for proteomics. Curr Opin Chem Biol, 12, 483–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansson O, Zetterberg H, Buchhave P, Londos E, Blennow K and Minthon L (2006) Association between CSF biomarkers and incipient Alzheimer’s disease in patients with mild cognitive impairment: a follow-up study. Lancet Neurol, 5, 228–234. [DOI] [PubMed] [Google Scholar]
- Hartl FU (2017) Protein Misfolding Diseases. Annu Rev Biochem, 86, 21–26. [DOI] [PubMed] [Google Scholar]
- Healy DG, Abou-Sleiman PM, Lees AJ, Casas JP, Quinn N, Bhatia K, Hingorani AD and Wood NW (2004) Tau gene and Parkinson’s disease: a case-control study and meta-analysis. J Neurol Neurosurg Psychiatry, 75, 962–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heller M, Mattou H, Menzel C and Yao X (2003) Trypsin catalyzed 16O-to-18O exchange for comparative proteomics: tandem mass spectrometry comparison using MALDI-TOF, ESI-QTOF, and ESI-ion trap mass spectrometers. J Am Soc Mass Spectrom, 14, 704–718. [DOI] [PubMed] [Google Scholar]
- Hendrickson RC, Lee AY, Song Q et al. (2015) High Resolution Discovery Proteomics Reveals Candidate Disease Progression Markers of Alzheimer’s Disease in Human Cerebrospinal Fluid. PLoS One, 10, e0135365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henriksen K, O’Bryant SE, Hampel H et al. (2014) The future of blood-based biomarkers for Alzheimer’s disease. Alzheimers Dement, 10, 115–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hertze J, Minthon L, Zetterberg H, Vanmechelen E, Blennow K and Hansson O (2010) Evaluation of CSF biomarkers as predictors of Alzheimer’s disease: a clinical follow-up study of 4.7 years. J Alzheimers Dis, 21, 1119–1128. [DOI] [PubMed] [Google Scholar]
- Hong Z, Shi M, Chung KA et al. (2010) DJ-1 and alpha-synuclein in human cerebrospinal fluid as biomarkers of Parkinson’s disease. Brain, 133, 713–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoofnagle AN and Wener MH (2009) The fundamental flaws of immunoassays and potential solutions using tandem mass spectrometry. J Immunol Methods, 347, 3–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horvatovich PL and Bischoff R (2010) Current technological challenges in biomarker discovery and validation. Eur J Mass Spectrom (Chichester), 16, 101–121. [DOI] [PubMed] [Google Scholar]
- Howitt J and Hill AF (2016) Exosomes in the Pathology of Neurodegenerative Diseases. The Journal of biological chemistry, 291, 26589–26597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hughes AJ, Daniel SE, Kilford L and Lees AJ (1992) Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. Journal of neurology, neurosurgery, and psychiatry, 55, 181–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hulstaert F, Blennow K, Ivanoiu A et al. (1999) Improved discrimination of AD patients using beta-amyloid(1–42) and tau levels in CSF. Neurology, 52, 1555–1562. [DOI] [PubMed] [Google Scholar]
- Hwang H, Zhang J, Chung KA et al. (2010) Glycoproteomics in neurodegenerative diseases. Mass Spectrom Rev, 29, 79–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inoue K, Hosaka D, Mochizuki N, Akatsu H, Tsutsumiuchi K, Hashizume Y, Matsukawa N, Yamamoto T and Toyo’oka T (2014) Simultaneous determination of post-translational racemization and isomerization of N-terminal amyloid-beta in Alzheimer’s brain tissues by covalent chiral derivatized ultraperformance liquid chromatography tandem mass spectrometry. Anal Chem, 86, 797–804. [DOI] [PubMed] [Google Scholar]
- Jack CR Jr., Petersen RC, Xu YC et al. (1999) Prediction of AD with MRI-based hippocampal volume in mild cognitive impairment. Neurology, 52, 1397–1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaffe JD, Keshishian H, Chang B, Addona TA, Gillette MA and Carr SA (2008) Accurate inclusion mass screening: a bridge from unbiased discovery to targeted assay development for biomarker verification. Mol. Cell. Proteomics, 7, 1952–1962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janelidze S, Stomrud E, Palmqvist S et al. (2016a) Plasma beta-amyloid in Alzheimer’s disease and vascular disease. Sci. Rep, 6, 26801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janelidze S, Zetterberg H, Mattsson N et al. (2016b) CSF Abeta42/Abeta40 and Abeta42/Abeta38 ratios: better diagnostic markers of Alzheimer disease. Ann Clin Transl Neurol, 3, 154–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeromin A and Bowser R (2017) Biomarkers in Neurodegenerative Diseases. Advances in neurobiology, 15, 491–528. [DOI] [PubMed] [Google Scholar]
- Jho YS, Zhulina EB, Kim MW and Pincus PA (2010) Monte carlo simulations of tau proteins: effect of phosphorylation. Biophys J, 99, 2387–2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin J, Hulette C, Wang Y, Zhang T, Pan C, Wadhwa R and Zhang J (2006) Proteomic identification of a stress protein, mortalin/mthsp70/GRP75: relevance to Parkinson disease. Mol Cell Proteomics, 5, 1193–1204. [DOI] [PubMed] [Google Scholar]
- Jin L, Wang D, Gooden DM, Ball CH and Fitzgerald MC (2016) Targeted Mass Spectrometry-Based Approach for Protein-Ligand Binding Analyses in Complex Biological Mixtures Using a Phenacyl Bromide Modification Strategy. Anal Chem, 88, 10987–10993. [DOI] [PubMed] [Google Scholar]
- Jouanne M, Rault S and Voisin-Chiret AS (2017) Tau protein aggregation in Alzheimer’s disease: An attractive target for the development of novel therapeutic agents. Eur J Med Chem, 139, 153–167. [DOI] [PubMed] [Google Scholar]
- Kalia LV and Kalia SK (2015) alpha-Synuclein and Lewy pathology in Parkinson’s disease. Curr Opin Neurol, 28, 375–381. [DOI] [PubMed] [Google Scholar]
- Kang JH, Irwin DJ, Chen-Plotkin AS et al. (2013) Association of cerebrospinal fluid beta-amyloid 1–42, T-tau, P-tau181, and alpha-synuclein levels with clinical features of drug-naive patients with early Parkinson disease. JAMA Neurol, 70, 1277–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitsou E, Pan S, Zhang J et al. (2008) Identification of proteins in human substantia nigra. Proteomics Clin Appl, 2, 776–782. [DOI] [PubMed] [Google Scholar]
- Kiyonami R, Schoen A, Prakash A, Peterman S, Zabrouskov V, Picotti P, Aebersold R, Huhmer A and Domon B (2011) Increased selectivity, analytical precision, and throughput in targeted proteomics. Molecular & cellular proteomics : MCP, 10, M110.002931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klatka LA, Schiffer RB, Powers JM and Kazee AM (1996) Incorrect diagnosis of Alzheimer’s disease. A clinicopathologic study. Arch Neurol, 53, 35–42. [DOI] [PubMed] [Google Scholar]
- Klein C and Westenberger A (2012) Genetics of Parkinson’s disease. Cold Spring Harb Perspect Med, 2, a008888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koivunen J, Scheinin N, Virta JR et al. (2010) Amyloid PET imaging in patients with mild cognitive impairment: a 2-year follow-up study. Neurology, 76, 1085–1090. [DOI] [PubMed] [Google Scholar]
- Konermann L, Pan J and Liu YH (2011) Hydrogen exchange mass spectrometry for studying protein structure and dynamics. Chem Soc Rev, 40, 1224–1234. [DOI] [PubMed] [Google Scholar]
- Korecka M, Waligorska T, Figurski M, Toledo JB, Arnold SE, Grossman M, Trojanowski JQ and Shaw LM (2014) Qualification of a surrogate matrix-based absolute quantification method for amyloid-beta(4)(2) in human cerebrospinal fluid using 2D UPLC-tandem mass spectrometry. J Alzheimers Dis, 41, 441–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korff A, Liu C, Ginghina C, Shi M, Zhang J and Alzheimer’s Disease Neuroimaging, I. (2013) alpha-Synuclein in cerebrospinal fluid of Alzheimer’s disease and mild cognitive impairment. J Alzheimers Dis, 36, 679–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kruse N, Persson S, Alcolea D et al. (2015) Validation of a quantitative cerebrospinal fluid alpha-synuclein assay in a European-wide interlaboratory study. Neurobiol. Aging, 36, 2587–2596. [DOI] [PubMed] [Google Scholar]
- Kuhlmann J, Andreasson U, Pannee J et al. (2017) CSF Abeta1–42 - an excellent but complicated Alzheimer’s biomarker - a route to standardisation. Clin Chim Acta, 467, 27–33. [DOI] [PubMed] [Google Scholar]
- Kuzyk MA, Parker CE, Domanski D and Borchers CH (2013) Development of MRM-based assays for the absolute quantitation of plasma proteins. Methods in molecular biology, 1023, 53–82. [DOI] [PubMed] [Google Scholar]
- Langen H, Takacs B, Evers S, Berndt P, Lahm HW, Wipf B, Gray C and Fountoulakis M (2000) Two-dimensional map of the proteome of Haemophilus influenzae. Electrophoresis, 21, 411–429. [DOI] [PubMed] [Google Scholar]
- Lashuel HA, Overk CR, Oueslati A and Masliah E (2013) The many faces of alpha-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci, 14, 38–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee PH, Lee G, Park HJ, Bang OY, Joo IS and Huh K (2006) The plasma alpha-synuclein levels in patients with Parkinson’s disease and multiple system atrophy. J Neural Transm (Vienna), 113, 1435–1439. [DOI] [PubMed] [Google Scholar]
- Lee TH, D’Asti E, Magnus N, Al-Nedawi K, Meehan B and Rak J (2011) Microvesicles as mediators of intercellular communication in cancer--the emerging science of cellular ‘debris’. Semin Immunopathol, 33, 455–467. [DOI] [PubMed] [Google Scholar]
- Lehnert S, Jesse S, Rist W et al. (2012) iTRAQ and multiple reaction monitoring as proteomic tools for biomarker search in cerebrospinal fluid of patients with Parkinson’s disease dementia. Exp Neurol, 234, 499–505. [DOI] [PubMed] [Google Scholar]
- Leinenbach A, Pannee J, Dulffer T et al. (2014) Mass spectrometry-based candidate reference measurement procedure for quantification of amyloid-beta in cerebrospinal fluid. Clin Chem, 60, 987–994. [DOI] [PubMed] [Google Scholar]
- Levitsky LI, Ivanov MV, Lobas AA and Gorshkov MV (2017) Unbiased False Discovery Rate Estimation for Shotgun Proteomics Based on the Target-Decoy Approach. J Proteome Res, 16, 393–397. [DOI] [PubMed] [Google Scholar]
- Lewczuk P, Matzen A, Blennow K, Parnetti L, Molinuevo JL, Eusebi P, Kornhuber J, Morris JC and Fagan AM (2017) Cerebrospinal Fluid Abeta42/40 Corresponds Better than Abeta42 to Amyloid PET in Alzheimer’s Disease. J Alzheimers Dis, 55, 813–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li QX, Mok SS, Laughton KM, McLean CA, Cappai R, Masters CL, Culvenor JG and Horne MK (2007) Plasma alpha-synuclein is decreased in subjects with Parkinson’s disease. Exp Neurol, 204, 583–588. [DOI] [PubMed] [Google Scholar]
- Lim A, Tsuang D, Kukull W et al. (1999) Clinico-neuropathological correlation of Alzheimer’s disease in a community-based case series. J Am Geriatr Soc, 47, 564–569. [DOI] [PubMed] [Google Scholar]
- Lin X, Cook TJ, Zabetian CP et al. (2012) DJ-1 isoforms in whole blood as potential biomarkers of Parkinson disease. Scientific reports, 2, 954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H, Sadygov RG and Yates JR 3rd (2004) A model for random sampling and estimation of relative protein abundance in shotgun proteomics. Anal Chem, 76, 4193–4201. [DOI] [PubMed] [Google Scholar]
- Loane C and Politis M (2011) Positron emission tomography neuroimaging in Parkinson’s disease. American journal of translational research, 3, 323–341. [PMC free article] [PubMed] [Google Scholar]
- Lovheim H, Elgh F, Johansson A, Zetterberg H, Blennow K, Hallmans G and Eriksson S (2017) Plasma concentrations of free amyloid beta cannot predict the development of Alzheimer’s disease. Alzheimers Dement, 13, 778–782. [DOI] [PubMed] [Google Scholar]
- Maita C, Tsuji S, Yabe I, Hamada S, Ogata A, Maita H, Iguchi-Ariga SM, Sasaki H and Ariga H (2008) Secretion of DJ-1 into the serum of patients with Parkinson’s disease. Neurosci Lett, 431, 86–89. [DOI] [PubMed] [Google Scholar]
- Makawita S and Diamandis EP (2010) The bottleneck in the cancer biomarker pipeline and protein quantification through mass spectrometry-based approaches: current strategies for candidate verification. Clin Chem, 56, 212–222. [DOI] [PubMed] [Google Scholar]
- Mallick P, Schirle M, Chen SS et al. (2007) Computational prediction of proteotypic peptides for quantitative proteomics. Nat Biotechnol, 25, 125–131. [DOI] [PubMed] [Google Scholar]
- Malm T, Loppi S and Kanninen KM (2016) Exosomes in Alzheimer’s disease. Neurochemistry international, 97, 193–199. [DOI] [PubMed] [Google Scholar]
- Marsili L, Rizzo G and Colosimo C (2018) Diagnostic Criteria for Parkinson’s Disease: From James Parkinson to the Concept of Prodromal Disease. Frontiers in neurology, 9, 156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA and Cummings JL (2015) Alzheimer’s disease. Nat Rev Dis Primers, 1, 15056. [DOI] [PubMed] [Google Scholar]
- Matsumoto J, Stewart T, Banks WA and Zhang J (2017a) The Transport Mechanism of Extracellular Vesicles at the Blood-Brain Barrier. Curr Pharm Des, 23, 6206–6214. [DOI] [PubMed] [Google Scholar]
- Matsumoto J, Stewart T, Sheng L, Li N, Bullock K, Song N, Shi M, Banks WA and Zhang J (2017b) Transmission of alpha-synuclein-containing erythrocyte-derived extracellular vesicles across the blood-brain barrier via adsorptive mediated transcytosis: another mechanism for initiation and progression of Parkinson’s disease? Acta neuropathologica communications, 5, 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mattsson N, Andreasson U, Persson S et al. (2011) The Alzheimer’s Association external quality control program for cerebrospinal fluid biomarkers. Alzheimers Dement, 7, 386–395.e386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mattsson N, Andreasson U, Persson S et al. (2013) CSF biomarker variability in the Alzheimer’s Association quality control program. Alzheimers Dement, 9, 251–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mattsson N, Zetterberg H, Hansson O et al. (2009) CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. JAMA, 302, 385–393. [DOI] [PubMed] [Google Scholar]
- Mayeux R (2004) Biomarkers: potential uses and limitations. NeuroRx, 1, 182–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDermott JE, Wang J, Mitchell H, Webb-Robertson BJ, Hafen R, Ramey J and Rodland KD (2013) Challenges in Biomarker Discovery: Combining Expert Insights with Statistical Analysis of Complex Omics Data. Expert Opin Med Diagn, 7, 37–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng Z and Veenstra TD (2011) Targeted mass spectrometry approaches for protein biomarker verification. Journal of proteomics, 74, 2650–2659. [DOI] [PubMed] [Google Scholar]
- Miller DB and O’Callaghan JP (2015) Biomarkers of Parkinson’s disease: present and future. Metabolism: clinical and experimental, 64, S40–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirgorodskaya OA, Kozmin YP, Titov MI, Korner R, Sonksen CP and Roepstorff P (2000) Quantitation of peptides and proteins by matrix-assisted laser desorption/ionization mass spectrometry using (18)O-labeled internal standards. Rapid Commun Mass Spectrom, 14, 1226–1232. [DOI] [PubMed] [Google Scholar]
- Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel-Doring F, Trenkwalder C and Schlossmacher MG (2011) alpha-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: a cohort study. Lancet Neurol, 10, 230–240. [DOI] [PubMed] [Google Scholar]
- Montine TJ, Shi M, Quinn JF et al. (2010) CSF Abeta(42) and tau in Parkinson’s disease with cognitive impairment. Mov Disord, 25, 2682–2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno-Gonzalo O, Villarroya-Beltri C and Sanchez-Madrid F (2014) Post-translational modifications of exosomal proteins. Front Immunol, 5, 383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris M, Maeda S, Vossel K and Mucke L (2011) The many faces of tau. Neuron, 70, 410–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Musiek ES and Holtzman DM (2015) Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat Neurosci, 18, 800–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakamura A, Kaneko N, Villemagne VL et al. (2018) High performance plasma amyloid-beta biomarkers for Alzheimer’s disease. Nature, 554, 249–254. [DOI] [PubMed] [Google Scholar]
- Neilson KA, Ali NA, Muralidharan S, Mirzaei M, Mariani M, Assadourian G, Lee A, van Sluyter SC and Haynes PA (2011) Less label, more free: approaches in label-free quantitative mass spectrometry. Proteomics, 11, 535–553. [DOI] [PubMed] [Google Scholar]
- Niemantsverdriet E, Ottoy J, Somers C et al. (2017) The Cerebrospinal Fluid Abeta1–42/Abeta1–40 Ratio Improves Concordance with Amyloid-PET for Diagnosing Alzheimer’s Disease in a Clinical Setting. J Alzheimers Dis, 60, 561–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nurmi E, Ruottinen HM, Bergman J, Haaparanta M, Solin O, Sonninen P and Rinne JO (2001) Rate of progression in Parkinson’s disease: a 6-[18F]fluoro-L-dopa PET study. Mov Disord, 16, 608–615. [DOI] [PubMed] [Google Scholar]
- Oda Y, Huang K, Cross FR, Cowburn D and Chait BT (1999) Accurate quantitation of protein expression and site-specific phosphorylation. Proc Natl Acad Sci U S A, 96, 6591–6596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oeckl P, Metzger F, Nagl M, von Arnim CA, Halbgebauer S, Steinacker P, Ludolph AC and Otto M (2016) 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, 15, 3126–3138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olanow CW and Stocchi F (2017) Levodopa: A new look at an old friend. Movement disorders : official journal of the Movement Disorder Society. [DOI] [PubMed] [Google Scholar]
- Olsson B, Lautner R, Andreasson U et al. (2016) CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol, 15, 673–684. [DOI] [PubMed] [Google Scholar]
- Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H, Pandey A and Mann M (2002) Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics, 1, 376–386. [DOI] [PubMed] [Google Scholar]
- Ono M, Shitashige M, Honda K, Isobe T, Kuwabara H, Matsuzuki H, Hirohashi S and Yamada T (2006) Label-free quantitative proteomics using large peptide data sets generated by nanoflow liquid chromatography and mass spectrometry. Mol Cell Proteomics, 5, 1338–1347. [DOI] [PubMed] [Google Scholar]
- Otsuki T, Wada H, Nakazawa N et al. (2001) Establishment of CD7+ human myeloma sister cell lines, KMS-21-PE and KMS-21-BM, carrying t(11;14) and t(8;14). Leuk Lymphoma, 42, 761–774. [DOI] [PubMed] [Google Scholar]
- Oueslati A, Fournier M and Lashuel HA (2010) Role of post-translational modifications in modulating the structure, function and toxicity of alpha-synuclein: implications for Parkinson’s disease pathogenesis and therapies. Progress in brain research, 183, 115–145. [DOI] [PubMed] [Google Scholar]
- Ovod V, Ramsey KN, Mawuenyega KG et al. (2017) Amyloid beta concentrations and stable isotope labeling kinetics of human plasma specific to central nervous system amyloidosis. Alzheimers Dement, 13, 841–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paleologou KE, Oueslati A, Shakked G et al. (2010) Phosphorylation at S87 is enhanced in synucleinopathies, inhibits alpha-synuclein oligomerization, and influences synuclein-membrane interactions. J Neurosci, 30, 3184–3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan C, Zhou Y, Dator R et al. (2014) Targeted discovery and validation of plasma biomarkers of Parkinson’s disease. J Proteome Res, 13, 4535–4545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pannee J, Portelius E, Oppermann M et al. (2013) A selected reaction monitoring (SRM)-based method for absolute quantification of Abeta38, Abeta40, and Abeta42 in cerebrospinal fluid of Alzheimer’s disease patients and healthy controls. J Alzheimers Dis, 33, 1021–1032. [DOI] [PubMed] [Google Scholar]
- Parker CE and Borchers CH (2014) Mass spectrometry based biomarker discovery, verification, and validation--quality assurance and control of protein biomarker assays. Mol Oncol, 8, 840–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parnetti L, Chiasserini D, Bellomo G et al. (2011) Cerebrospinal fluid Tau/alpha-synuclein ratio in Parkinson’s disease and degenerative dementias. Mov Disord, 26, 1428–1435. [DOI] [PubMed] [Google Scholar]
- Parnetti L, Farotti L, Eusebi P et al. (2014) Differential role of CSF alpha-synuclein species, tau, and Abeta42 in Parkinson’s Disease. Front Aging Neurosci, 6, 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paterson RW, Heywood WE, Heslegrave AJ et al. (2016) A targeted proteomic multiplex CSF assay identifies increased malate dehydrogenase and other neurodegenerative biomarkers in individuals with Alzheimer’s disease pathology. Transl Psychiatry, 6, e952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paulovich AG, Whiteaker JR, Hoofnagle AN and Wang P (2008) The interface between biomarker discovery and clinical validation: The tar pit of the protein biomarker pipeline. Proteomics Clin Appl, 2, 1386–1402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pearl GS (1997) Diagnosis of Alzheimer’s disease in a community hospital-based brain bank program. South Med J, 90, 720–722. [DOI] [PubMed] [Google Scholar]
- Peng M, Taouatas N, Cappadona S, van Breukelen B, Mohammed S, Scholten A and Heck AJ (2012) Protease bias in absolute protein quantitation. Nat Methods, 9, 524–525. [DOI] [PubMed] [Google Scholar]
- Peterson AC, Russell JD, Bailey DJ, Westphall MS and Coon JJ (2012) Parallel reaction monitoring for high resolution and high mass accuracy quantitative, targeted proteomics. Mol Cell Proteomics, 11, 1475–1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piccini P and Whone A (2004) Functional brain imaging in the differential diagnosis of Parkinson’s disease. Lancet Neurol, 3, 284–290. [DOI] [PubMed] [Google Scholar]
- Picotti P and Aebersold R (2012) Selected reaction monitoring-based proteomics: workflows, potential, pitfalls and future directions. Nature methods, 9, 555–566. [DOI] [PubMed] [Google Scholar]
- Podlesniy P, Figueiro-Silva J, Llado A et al. (2013) Low cerebrospinal fluid concentration of mitochondrial DNA in preclinical Alzheimer disease. Annals of neurology, 74, 655–668. [DOI] [PubMed] [Google Scholar]
- Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE and Lang AE (2017) Parkinson disease. Nat Rev Dis Primers, 3, 17013. [DOI] [PubMed] [Google Scholar]
- Poste G (2011) Bring on the biomarkers. Nature, 469, 156–157. [DOI] [PubMed] [Google Scholar]
- Pottiez G, Yang L, Stewart T et al. (2017) Mass-Spectrometry-Based Method To Quantify in Parallel Tau and Amyloid beta 1–42 in CSF for the Diagnosis of Alzheimer’s Disease. J Proteome Res, 16, 1228–1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Price JL, Ko AI, Wade MJ, Tsou SK, McKeel DW and Morris JC (2001) Neuron number in the entorhinal cortex and CA1 in preclinical Alzheimer disease. Arch Neurol, 58, 1395–1402. [DOI] [PubMed] [Google Scholar]
- Puntmann VO (2009) How-to guide on biomarkers: biomarker definitions, validation and applications with examples from cardiovascular disease. Postgraduate medical journal, 85, 538–545. [DOI] [PubMed] [Google Scholar]
- Ranginwala NA, Hynan LS, Weiner MF and White CL 3rd (2008) Clinical criteria for the diagnosis of Alzheimer disease: still good after all these years. Am J Geriatr Psychiatry, 16, 384–388. [DOI] [PubMed] [Google Scholar]
- Rauniyar N (2015) Parallel Reaction Monitoring: A Targeted Experiment Performed Using High Resolution and High Mass Accuracy Mass Spectrometry. Int J Mol Sci, 16, 28566–28581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rembach A, Faux NG, Watt AD et al. (2014) Changes in plasma amyloid beta in a longitudinal study of aging and Alzheimer’s disease. Alzheimers Dement, 10, 53–61. [DOI] [PubMed] [Google Scholar]
- Rifai N, Gillette MA and Carr SA (2006) Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat Biotechnol, 24, 971–983. [DOI] [PubMed] [Google Scholar]
- Risacher SL and Saykin AJ (2013) Neuroimaging biomarkers of neurodegenerative diseases and dementia. Semin Neurol, 33, 386–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ritchie C, Smailagic N, Noel-Storr AH, Ukoumunne O, Ladds EC and Martin S (2017) CSF tau and the CSF tau/ABeta ratio for the diagnosis of Alzheimer’s disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev, 3, CD010803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson PA (2008) Protein stability and aggregation in Parkinson’s disease. Biochem J, 413, 1–13. [DOI] [PubMed] [Google Scholar]
- Ronsein GE, Pamir N, von Haller PD, Kim DS, Oda MN, Jarvik GP, Vaisar T and Heinecke JW (2015) Parallel reaction monitoring (PRM) and selected reaction monitoring (SRM) exhibit comparable linearity, dynamic range and precision for targeted quantitative HDL proteomics. J Proteomics, 113, 388–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ross PL, Huang YN, Marchese JN et al. (2004) Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol Cell Proteomics, 3, 1154–1169. [DOI] [PubMed] [Google Scholar]
- Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ and Pereira de Almeida L (2017) Extracellular vesicles: Novel promising delivery systems for therapy of brain diseases. Journal of controlled release : official journal of the Controlled Release Society, 262, 247–258. [DOI] [PubMed] [Google Scholar]
- Sauvee M, DidierLaurent G, Latarche C, Escanye MC, Olivier JL and Malaplate-Armand C (2014) Additional use of Abeta(4)(2)/Abeta(4)(0) ratio with cerebrospinal fluid biomarkers P-tau and Abeta(4)(2) increases the level of evidence of Alzheimer’s disease pathophysiological process in routine practice. J Alzheimers Dis, 41, 377–386. [DOI] [PubMed] [Google Scholar]
- Schapira AH (2013) Recent developments in biomarkers in Parkinson disease. Curr. Opin. Neurol, 26, 395–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schilling LP, Zimmer ER, Shin M et al. (2016) Imaging Alzheimer’s disease pathophysiology with PET. Dement Neuropsychol, 10, 79–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmid AW, Fauvet B, Moniatte M and Lashuel HA (2013) Alpha-synuclein post-translational modifications as potential biomarkers for Parkinson disease and other synucleinopathies. Molecular & cellular proteomics : MCP, 12, 3543–3558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selevsek N, Matondo M, Sanchez Carbayo M, Aebersold R and Domon B (2011) Systematic quantification of peptides/proteins in urine using selected reaction monitoring. Proteomics, 11, 1135–1147. [DOI] [PubMed] [Google Scholar]
- Semla TP (2007) Alzheimer’s disease: will advances made in the past turn the tide? Ann. Pharmacother, 41, 1494–1496. [DOI] [PubMed] [Google Scholar]
- Shah P, Cho SK, Thulstrup PW, Bjerrum MJ, Lee PH, Kang JH, Bhang YJ and Yang SW (2017) MicroRNA Biomarkers in Neurodegenerative Diseases and Emerging Nano-Sensors Technology. Journal of movement disorders, 10, 18–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shahnawaz M, Tokuda T, Waragai M, Mendez N, Ishii R, Trenkwalder C, Mollenhauer B and Soto C (2017) Development of a Biochemical Diagnosis of Parkinson Disease by Detection of alpha-Synuclein Misfolded Aggregates in Cerebrospinal Fluid. JAMA Neurol, 74, 163–172. [DOI] [PubMed] [Google Scholar]
- Shaw LM, Vanderstichele H, Knapik-Czajka M et al. (2009) Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann Neurol, 65, 403–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheinerman KS, Toledo JB, Tsivinsky VG et al. (2017) Circulating brain-enriched microRNAs as novel biomarkers for detection and differentiation of neurodegenerative diseases. Alzheimer’s research & therapy, 9, 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi M, Bradner J, Hancock AM et al. (2011) Cerebrospinal fluid biomarkers for Parkinson disease diagnosis and progression. Ann Neurol, 69, 570–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi M, Kovac A, Korff A et al. (2016a) CNS tau efflux via exosomes is likely increased in Parkinson’s disease but not in Alzheimer’s disease. Alzheimers Dement, 12, 1125–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi M, Kovac A, Korff A et al. (2016b) CNS tau efflux via exosomes is likely increased in Parkinson disease but not in Alzheimer disease. Alzheimers Dement, 10.1016/j.jalz.2016.1004.1003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi M, Liu C, Cook TJ et al. (2014) Plasma exosomal alpha-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol, 128, 639–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi M, Movius J, Dator R et al. (2015) Cerebrospinal fluid peptides as potential Parkinson disease biomarkers: a staged pipeline for discovery and validation. Mol Cell Proteomics, 14, 544–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi M, Zabetian CP, Hancock AM et al. (2010) Significance and confounders of peripheral DJ-1 and alpha-synuclein in Parkinson’s disease. Neurosci Lett, 480, 78–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi T, Fillmore TL, Sun X et al. (2012) Antibody-free, targeted mass-spectrometric approach for quantification of proteins at low picogram per milliliter levels in human plasma/serum. Proc Natl Acad Sci U S A, 109, 15395–15400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siderowf A, Xie SX, Hurtig H et al. (2010) CSF amyloid {beta} 1–42 predicts cognitive decline in Parkinson disease. Neurology, 75, 1055–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simon-Sanchez J, Schulte C, Bras JM et al. (2009) Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat. Genet, 41, 1308–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skogseth RE, Bronnick K, Pereira JB, Mollenhauer B, Weintraub D, Fladby T and Aarsland D (2015) Associations between Cerebrospinal Fluid Biomarkers and Cognition in Early Untreated Parkinson’s Disease. J Parkinsons Dis, 5, 783–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Snyder HM, Carrillo MC, Grodstein F et al. (2014) Developing novel blood-based biomarkers for Alzheimer’s disease. Alzheimers Dement, 10, 109–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldan A, Pettigrew C, Cai Q, Wang MC, Moghekar AR, O’Brien RJ, Selnes OA and Albert MS (2016) Hypothetical Preclinical Alzheimer Disease Groups and Longitudinal Cognitive Change. JAMA Neurol, 73, 698–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song F, Poljak A, Smythe GA and Sachdev P (2009) Plasma biomarkers for mild cognitive impairment and Alzheimer’s disease. Brain Res. Rev, 61, 69–80. [DOI] [PubMed] [Google Scholar]
- Spellman DS, Wildsmith KR, Honigberg LA et al. (2015) Development and evaluation of a multiplexed mass spectrometry based assay for measuring candidate peptide biomarkers in Alzheimer’s Disease Neuroimaging Initiative (ADNI) CSF. Proteomics Clin Appl, 9, 715–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stewart T, Sossi V, Aasly JO et al. (2015) Phosphorylated alpha-synuclein in Parkinson’s disease: correlation depends on disease severity. Acta Neuropathol Commun, 3, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strimbu K and Tavel JA (2010) What are biomarkers? Curr Opin HIV AIDS, 5, 463–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Surinova S, Schiess R, Huttenhain R, Cerciello F, Wollscheid B and Aebersold R (2011) On the development of plasma protein biomarkers. J Proteome Res, 10, 5–16. [DOI] [PubMed] [Google Scholar]
- Swaminathan S, Risacher SL, Yoder KK et al. (2014) Association of plasma and cortical amyloid beta is modulated by APOE epsilon4 status. Alzheimers Dement, 10, e9–e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sweeney P, Park H, Baumann M et al. (2017) Protein misfolding in neurodegenerative diseases: implications and strategies. Transl Neurodegener, 6, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takahashi RH, Milner TA, Li F et al. (2002) Intraneuronal Alzheimer abeta42 accumulates in multivesicular bodies and is associated with synaptic pathology. The American journal of pathology, 161, 1869–1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang BL and Kumar R (2008) Biomarkers of mild cognitive impairment and Alzheimer’s disease. Ann. Acad. Med. Singapore, 37, 406–410. [PubMed] [Google Scholar]
- Terrelonge M Jr., Marder KS, Weintraub D and Alcalay RN (2016) CSF beta-Amyloid 1–42 Predicts Progression to Cognitive Impairment in Newly Diagnosed Parkinson Disease. J Mol Neurosci, 58, 88–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thambisetty M and Lovestone S (2010) Blood-based biomarkers of Alzheimer’s disease: challenging but feasible. Biomark Med, 4, 65–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thompson A, Schafer J, Kuhn K et al. (2003) Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Anal Chem, 75, 1895–1904. [DOI] [PubMed] [Google Scholar]
- Thompson AG, Gray E, Heman-Ackah SM, Mager I, Talbot K, Andaloussi SE, Wood MJ and Turner MR (2016) Extracellular vesicles in neurodegenerative disease - pathogenesis to biomarkers. Nat. Rev. Neurol, 12, 346–357. [DOI] [PubMed] [Google Scholar]
- Tokuda T, Salem SA, Allsop D, Mizuno T, Nakagawa M, Qureshi MM, Locascio JJ, Schlossmacher MG and El-Agnaf OM (2006) Decreased alpha-synuclein in cerebrospinal fluid of aged individuals and subjects with Parkinson’s disease. Biochem Biophys Res Commun, 349, 162–166. [DOI] [PubMed] [Google Scholar]
- Tolosa E, Wenning G and Poewe W (2006) The diagnosis of Parkinson’s disease. The Lancet. Neurology, 5, 75–86. [DOI] [PubMed] [Google Scholar]
- Trojanowski JQ, Vandeerstichele H, Korecka M et al. (2010) Update on the biomarker core of the Alzheimer’s Disease Neuroimaging Initiative subjects. Alzheimer’s & dementia : the journal of the Alzheimer’s Association, 6, 230–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsiatsiani L and Heck AJ (2015) Proteomics beyond trypsin. The FEBS journal, 282, 2612–2626. [DOI] [PubMed] [Google Scholar]
- Tzoulis C, Tran GT, Schwarzlmuller T et al. (2013) Severe nigrostriatal degeneration without clinical parkinsonism in patients with polymerase gamma mutations. Brain, 136, 2393–2404. [DOI] [PubMed] [Google Scholar]
- Vidova V and Spacil Z (2017) A review on mass spectrometry-based quantitative proteomics: Targeted and data independent acquisition. Analytica chimica acta, 964, 7–23. [DOI] [PubMed] [Google Scholar]
- Vosseller K (2007) Proteomics of Alzheimer’s disease: Unveiling protein dysregulation in complex neuronal systems. Proteomics Clin Appl, 1, 1351–1361. [DOI] [PubMed] [Google Scholar]
- Wang PN, Lirng JF, Lin KN, Chang FC and Liu HC (2006) Prediction of Alzheimer’s disease in mild cognitive impairment: a prospective study in Taiwan. Neurobiol. Aging, 27, 1797–1806. [DOI] [PubMed] [Google Scholar]
- Wang Y, Shi M, Chung KA et al. (2012) Phosphorylated alpha-synuclein in Parkinson’s disease. Sci Transl Med, 4, 121ra120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waragai M, Nakai M, Wei J et al. (2007) Plasma levels of DJ-1 as a possible marker for progression of sporadic Parkinson’s disease. Neurosci Lett, 425, 18–22. [DOI] [PubMed] [Google Scholar]
- Waragai M, Wei J, Fujita M, Nakai M, Ho GJ, Masliah E, Akatsu H, Yamada T and Hashimoto M (2006) Increased level of DJ-1 in the cerebrospinal fluids of sporadic Parkinson’s disease. Biochem Biophys Res Commun, 345, 967–972. [DOI] [PubMed] [Google Scholar]
- Weiner MW, Veitch DP, Aisen PS et al. (2012) The Alzheimer’s Disease Neuroimaging Initiative: a review of papers published since its inception. Alzheimer’s & dementia : the journal of the Alzheimer’s Association, 8, S1–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West MJ, Kawas CH, Stewart WF, Rudow GL and Troncoso JC (2004) Hippocampal neurons in pre-clinical Alzheimer’s disease. Neurobiol. Aging, 25, 1205–1212. [DOI] [PubMed] [Google Scholar]
- Whiteaker JR, Lin C, Kennedy J et al. (2011) A targeted proteomics-based pipeline for verification of biomarkers in plasma. Nat. Biotechnol, 29, 625–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiener MC, Sachs JR, Deyanova EG and Yates NA (2004) Differential mass spectrometry: a label-free LC-MS method for finding significant differences in complex peptide and protein mixtures. Anal Chem, 76, 6085–6096. [DOI] [PubMed] [Google Scholar]
- Wildsmith KR, Schauer SP, Smith AM, Arnott D, Zhu Y, Haznedar J, Kaur S, Mathews WR and Honigberg LA (2014) Identification of longitudinally dynamic biomarkers in Alzheimer’s disease cerebrospinal fluid by targeted proteomics. Mol Neurodegener, 9, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Witkowska HE, Hall SC and Fisher SJ (2012) Breaking the bottleneck in the protein biomarker pipeline. Clin Chem, 58, 321–323. [DOI] [PubMed] [Google Scholar]
- Yang L, Stewart T, Shi M et al. (2017) An alpha-synuclein MRM assay with diagnostic potential for Parkinson’s disease and monitoring disease progression. Proteomics Clin Appl, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao X, Afonso C and Fenselau C (2003) Dissection of proteolytic 18O labeling: endoprotease-catalyzed 16O-to-18O exchange of truncated peptide substrates. J Proteome Res, 2, 147–152. [DOI] [PubMed] [Google Scholar]
- Zabetian CP, Hutter CM, Factor SA et al. (2007) Association analysis of MAPT H1 haplotype and subhaplotypes in Parkinson’s disease. Ann. Neurol, 62, 137–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zetterberg H (2015) Cerebrospinal fluid biomarkers for Alzheimer’s disease: current limitations and recent developments. Curr Opin Psychiatry, 28, 402–409. [DOI] [PubMed] [Google Scholar]
- Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J and Hansson O (2013) Plasma tau levels in Alzheimer’s disease. Alzheimers Res Ther, 5, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Song Y, Chen H and Fan D (2005) The tau gene haplotype h1 confers a susceptibility to Parkinson’s disease. Eur Neurol, 53, 15–21. [DOI] [PubMed] [Google Scholar]
- Zhang X, Li L, Wei D, Yap Y and Chen F (2007) Moving cancer diagnostics from bench to bedside. Trends in biotechnology, 25, 166–173. [DOI] [PubMed] [Google Scholar]
- Zheng T, Pu J, Chen Y et al. (2017) Plasma Exosomes Spread and Cluster Around beta-Amyloid Plaques in an Animal Model of Alzheimer’s Disease. Frontiers in aging neuroscience, 9, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziegler A, Koch A, Krockenberger K and Grosshennig A (2012) Personalized medicine using DNA biomarkers: a review. Hum Genet, 131, 1627–1638. [DOI] [PMC free article] [PubMed] [Google Scholar]




