Skip to content
Publicly Available Published by De Gruyter February 18, 2015

Quantitative detection of amyloid-β peptides by mass spectrometry: state of the art and clinical applications

  • Pauline Bros , Vincent Delatour EMAIL logo , Jérôme Vialaret , Béatrice Lalere , Nicolas Barthelemy , Audrey Gabelle , Sylvain Lehmann EMAIL logo and Christophe Hirtz

Abstract

Alzheimer’s disease (AD) is the most common form of dementia in humans, and a major public health concern with 35 million of patients worldwide. Cerebrospinal fluid (CSF) biomarkers being early diagnostic indicators of AD, it is essential to use the most efficient analytical methods to detect and quantify them accurately. These biomarkers, and more specifically amyloid-β (Aβ) peptides, are measured in routine clinical practice using immunoassays. However, there are several limits to this immunodetection in terms of specificity and multiplexing of the multiple isoforms of the Aβ peptides. To overcome these issues, the quantification of these analytes by mass spectrometry (MS) represents an interesting alternative, and several assays have been described over the past years. This article reviews the different Aβ peptides quantitative MS-based approaches published so far, compares their pre-analytical phase, and the different quantitative strategies implemented that might be suitable for clinical applications.

Introduction

Alzheimer’s disease (AD) is the most common form of dementia, a general term encompassing loss of memory and cognitive functions interfering therefore with activities of daily living. With 35 million of patients worldwide, AD represents 50%–80% of cases of dementia and is reaching epidemic proportions in industrialized countries mainly because of the aging of the population. By 2050, the estimates are quite pessimistic with more than 115.4 million people expected to develop AD [1]. Two main forms of AD coexist, sporadic AD which affects around 5% of the population after the age of 65, and familial AD, which is rare (<1% of the patients), often appears before the age of 65, and is linked to mutations in AD relevant genes (PSEN1-2, APP) [2]. Regardless of its form, AD is characterized by behavioral changes, memory impairments, praxis and gnosis disorders leading to a progressive loss of autonomy in patients [3]. As for today, there are no curative treatments available, but patients can still benefit form symptomatic treatments which improve their quality of life [4].

On a neuro-pathological level, AD is characterized by the presence in the brain parenchyma of hyperphosphorylated tau (p-tau) proteins aggregated into neurofibrillary tangles, and amyloid plaques [5]. Amyloid plaques are formed by aggregates of amyloid-β (Aβ) peptides; the Aβ 1-42 isoform being the most represented in the plaques of autopsied brains in AD patients [6]. Aβ peptides derive from the amyloid precursor protein (APP), a 87-kDa transmembrane protein with a large extracellular domain. This protein is metabolically cleaved by α-, β- and γ-secretases, leading to the formation of several peptide fragments including Aβ 1-42 and 1-40 (Figure 1). The decrease of Aβ peptide concentrations in the cerebrospinal fluid (CSF) of AD patients is correlated with the presence of amyloid plaques [7]. This is especially true for Aβ 1-42 which has a higher tendency to aggregation than Aβ 1-40 [8]. Aβ 1-40 is therefore rather involved in advanced stages of the disease [9].

Figure 1: Metabolism of the amyloid precursor protein (APP).Graphic representation of the generation of Aβ peptide isoforms from the amyloid precursor protein (APP). APP can be metabolized in two main ways: the amyloid pathway and non-amyloid pathway. In the amyloid pathway, the action of a β-secretase followed by a γ-secretase generates the soluble APP-β, the classical Aβ peptides (1-42, 1-40, 1-38) and the amyloid intra cellular domain (AICD). The non-amyloid pathway via the α-secretase and the γ-secretase cleaves the APP in three fragments: the soluble APP-α, p3 and the AICD. Short truncated isoforms (Aβ 1-14, 1-16, 1-17) could result from an additional pathway combining the action of β-and α-secretases.
Figure 1:

Metabolism of the amyloid precursor protein (APP).

Graphic representation of the generation of Aβ peptide isoforms from the amyloid precursor protein (APP). APP can be metabolized in two main ways: the amyloid pathway and non-amyloid pathway. In the amyloid pathway, the action of a β-secretase followed by a γ-secretase generates the soluble APP-β, the classical Aβ peptides (1-42, 1-40, 1-38) and the amyloid intra cellular domain (AICD). The non-amyloid pathway via the α-secretase and the γ-secretase cleaves the APP in three fragments: the soluble APP-α, p3 and the AICD. Short truncated isoforms (Aβ 1-14, 1-16, 1-17) could result from an additional pathway combining the action of β-and α-secretases.

The diagnosis of AD is based on neuropsychological tests, functional and morphologic imaging (MRI, hippocampal volumetry), and the levels of the following biomarkers present in the CSF: Aβ peptides, tau and p-tau(181) proteins [10–13]. These biomarkers which are now routinely measured in specialized laboratories [12, 14] are useful to predict and diagnose AD. In practice, two Aβ peptides (Aβ 1-40 and Aβ 1-42) are quantified by enzyme-linked immuno-sorbent assay (ELISA) [13, 14]. This quantification is based the recognition of the epitopes corresponding to the C-terminus amino acids of Aβ 1-40 and Aβ 1-42 using, respectively, the 2G3 and 21F12 antibodies. Recently, new multiplex immunoassays detecting multiples isoforms of Aβ peptides (1-38, 1-40 and 1-42) have been developed using Meso Scale Discovery (MSD) and Luminex platforms [15–19].

Importantly, other Aβ peptides like Aβ 2-14, 1-17, 1-18, 1-33, 1-34 or 1-37 have been described [20, 21] and their role and diagnostic interest still need to be explored in human CSF. Developing multiplexed ELISA in this context would be lengthy and costly. Mass spectrometry (MS) with its high specificity and multiplexing capacity appears as a good alternative to immunoassay techniques, and could represent an interesting new diagnostic tool for AD. Incidentally, it is interesting to note that the number of publications related to the analysis of Aβ peptides by MS has increased 12-fold in the past 20 years.

Physicochemical properties of Aβ peptides

The origin of amyloid plaques is in relation with the tendency of Aβ peptides to aggregate, especially Aβ 1-42 [22]. Aβ peptide aggregation is mainly triggered by conformational changes [23]. These peptides which are initially part of the APP transmembrane domain acquire after proteolytic cleavage a β-sheet structure. Then, they aggregate in an antiparallel arrangement to form filaments. Several filaments constitute fibrils which eventually form amyloid plaques [23]. While Aβ peptides auto-aggregate, they also tend to bind to other proteins in vivo [24] and they also adsorb in a non-specific manner to the plastic of collection and assay tubes [25] rendering more complex their accurate quantification. The particular physicochemical properties of Aβ peptides require therefore to control and standardize pre-analytical and analytical phases to obtain reliable measurements with both MS and ELISA, and to eventually deliver reliable results to patients [26–28].

Pre-analytical conditions

Time of CSF collection

It is known that the concentration of some analytes in the CSF, like melatonin or dopamine, is influenced by circadian rhythms [29]. In 2007, Bateman et al. reported a significant daytime variation (1.5–6-fold) of Aβ 1-40 and Aβ 1-42 levels in CSF over a 36-h period [30]. However, several other studies did not observe any significant daytime variations [27, 31]. To be on the safe side, it is however recommended to always perform CSF collection at the same time [26].

CSF collection tubes

Composition of tubes used to collect CSF during lumbar puncture has a strong influence on Aβ peptide concentrations [32]. This is in relation with the tendency of Aβ peptides to adsorb to the walls of the tube. Hence, it has been shown that glass and polystyrene tubes led to lower measured concentrations of Aβ peptides [25, 31]. Polypropylene tubes were believed to reduce this phenomenon [32]. However, even polypropylene tubes which are not made entirely of polypropylene, and have various surface treatments, have a major impact on Aβ peptide measured concentrations [25]. The consequences on AD biological diagnosis could be very important, as demonstrated in a multisite study conducted in four French Centers for Memory Research and Resources (CMRR) (Montpellier, Lille, Lyon and Paris) [33]. Standardization of the collection tubes (using, e.g., the tube Sarstedt 62.610.201) could improve multicenter comparison and decrease the risk of wrong diagnosis [34].

CSF sample storage

Storage at –20 °C or –80 °C has apparently little influence on CSF Aβ peptides [31]. It is however recommended to freeze and store Aβ standards and CSF at –80 °C to avoid stability issues [35–37]. Addition of protease inhibitors could improve protein stability upon storage but add an additional complexity to samples that could affect their commutability [37]. As for many analytes, more than two or three freeze/thaw cycles have also to be avoided to ensure the reproducibility of the results [27].

Analytical strategies for the detection and the quantification of Aβ peptides by MS

As a result of their low abundance and the wide dynamic range of protein concentrations in CSF (1010) [38], Aβ peptides are difficult to detect by MS. A sample preparation step is therefore generally necessary in order to extract and concentrate them before analysis [39]. After thawing CSF, in order to recover monomeric Aβ peptides that otherwise spontaneously auto-aggregate, aggregate to other proteins or get adsorbed to the walls of tubes, the most common approach is to denature samples with guanidine hydrochloride (Gnd HCl 5M). This can also help minimizing enzymatic activities and thus improving intermediate precision of measurements by reducing sample degradation [40]. After denaturation, the samples can be analyzed by various MS techniques as described below and Table 1.

Table 1:

Detection of amyloid-β peptides.

ELISAMALDISELDILC-MS/MS
1-38, 1-40, 1-42xxxx
1-13, 1-14, 1-15,1-16, 1-20, 1-28, 1-30, 1-37 ox, 1-38 ox, 1-39 ox, 1-40 ox, 1-42 ox, 1-43, 2-42, 3-40, 3-42, 5-40xx
1-17, 1-18, 1-19, 1-33, 1-34, 1-39xxx
1-29, 2-15,2-16,2-17, 2-19, 2-38, 4-13, 4-14, 4-15, 4-16, 4-19x
1-35, 1-36, 1-37, 1-40 x-x, 3-44, 3-47, 10-40, 11-40x
2-14xx
4-40, 4-42, 5-42, 7-42, 8-42, 9-40, 9-42, 10-42, pGlu 11-42, pGlu 3-40, pGlu 3-42x
11-42xx

Immuno-precipitation – matrix-assisted laser desorption ionization-time of flight-MS (IP-MALDI-ToF-MS)

MALDI-ToF-MS is a sensitive and fast technique for the detection and relative quantification of Aβ peptides, yielding a relatively high resolution and high mass accuracy. MALDI is based on the use of a laser beam for desorption, sublimation and ionization of Aβ peptides co-crystalized with a chemical matrix (α-cyano-4-hydroxycinnamic acid) on a stainless steel target [41] (Figure 2A). However, due to the complex nature of CSF, extraction and purification steps, like immuno-precipitation (IP), are necessary to reduce the matrix complexity in order to measure Aβ peptides with sufficient sensitivity [43]. Immuno-capture techniques, like those described by Portelius et al., are based on magnetic beads (Dynabeads M-280 streptavidin) functionalized with anti-Aβ peptide antibodies. The most commonly used antibodies are 6E10 and 4G8 which are directed against epitopes 4-9 and 18-22, respectively [41, 44, 45]. As the 6E10 epitope is located in the N-terminus of the Aβ peptide, it captures mainly different truncated C-terminal isoforms. In 2006 and 2007, Portelius et al. detected 18 Aβ peptides truncated at the N-terminus after 6E10 IP and analysis on a Bruker Autoflex mass spectrometer [41, 46]. Interestingly, the authors showed that Aβ 1-16, Aβ 1-33, Aβ 1-39, and Aβ 1-42 expression profile in CSF (Table 1) could help differentiating patients with sporadic AD from non-demented controls with a global accuracy of 86% [41].

Figure 2: Illustration of MS detection approaches and profile of Aβ peptides.(A) MALDI-ToF-MS is a very sensitive technique to determine the mass of proteins and peptides. It is relatively fast and easy to use. It relies on a soft ionization technique in which a short laser pulse is used to ionize molecules. A protein or peptide sample is placed on a target plate where it is mixed with an appropriate chemical matrix. The co-crystallized mixture of samples and matrix is irradiated with a short laser pulse and entered in a gas phase. The ions are accelerated in a time of flight (ToF) analyzer which measures the time it takes for the molecules to travel a set distance. The mass to charge ratio (m/z) of the ions can then be calculated. (B) selected reaction monitoring (SRM) is a very specific and sensitive targeted MS technique used to quantify preselected relevant ions. It is based on the use of triple-quadrupole-type mass spectrometers coupled to HPLC. After peptide ionization in the source, one or several peptides with a known mass are selected in the first quadrupole. These “precursor” ions are then fragmented in the second quadrupole. The third quadrupole can monitor one or several ion fragments called “product” ions which are specific to the precursor ion. The monitoring of a precursor ion and one of its product ions is called a “transition” which is specific to the selected peptide and can be quantified. (C) Chromatogram obtained after SPE CSF fractionation according to the Pannee et al. protocol [42] and nanoLC-triple quadrupole analysis with and alkaline mobile phase.
Figure 2:

Illustration of MS detection approaches and profile of Aβ peptides.

(A) MALDI-ToF-MS is a very sensitive technique to determine the mass of proteins and peptides. It is relatively fast and easy to use. It relies on a soft ionization technique in which a short laser pulse is used to ionize molecules. A protein or peptide sample is placed on a target plate where it is mixed with an appropriate chemical matrix. The co-crystallized mixture of samples and matrix is irradiated with a short laser pulse and entered in a gas phase. The ions are accelerated in a time of flight (ToF) analyzer which measures the time it takes for the molecules to travel a set distance. The mass to charge ratio (m/z) of the ions can then be calculated. (B) selected reaction monitoring (SRM) is a very specific and sensitive targeted MS technique used to quantify preselected relevant ions. It is based on the use of triple-quadrupole-type mass spectrometers coupled to HPLC. After peptide ionization in the source, one or several peptides with a known mass are selected in the first quadrupole. These “precursor” ions are then fragmented in the second quadrupole. The third quadrupole can monitor one or several ion fragments called “product” ions which are specific to the precursor ion. The monitoring of a precursor ion and one of its product ions is called a “transition” which is specific to the selected peptide and can be quantified. (C) Chromatogram obtained after SPE CSF fractionation according to the Pannee et al. protocol [42] and nanoLC-triple quadrupole analysis with and alkaline mobile phase.

To take into account sample preparation variability, as well as differences in desorption/ionization, the area of each MS peak obtained for the Aβ peptides was normalized by the sum of peak areas of the most abundant Aβ peptides [41]. However, this approach, called internal normalization, does not yield information on the absolute concentration of Aβ peptides. It needs also to perform several successive MS acquisitions in order to obtain an interpretable profile for each sample. To overcome these issues, a quantitative analysis by isotope dilution has been developed. The previously described IP-MALDI-ToF-MS technique was modified by adding two isotopically-labeled Aβ peptides as internal standards, Aβ 1-15 Arg13C15N and Aβ 1-34 Arg13C15N [46]. These labeled peptides triggered a 10 Da shift of the nominal mass and thus did not interfere with MS peaks of endogenous peptides to be quantified. This resulted in an improvement in results accuracy for Aβ 1-17 and Aβ 1-40 peptides.

Of note, IP-MALDI-ToF MS was used to follow the effect of a γ-secretase inhibitor [Semagacestat (LY450139)] [47]. This compound resulted in an increased production of Aβ 1-14, 1-15, and 1-16, a decrease in Aβ 1-17 while Aβ 1-42 and 1-40 remained unaffected. This illustrates the interest of following additional Aβ isoforms in AD research [39, 48–51].

Of note, SELDI-ToF-MS which is based on an initial capture of Aβ peptides using antibodies covalently immobilized on a proteinchip prior to MALDI-ToF has been used in the past to detect different isoforms of these analytes [52]. This straightforward approach allowed the discovery of several new AD biomarkers [20, 53] and in particular two new Aβ peptides (Aβ 3-44, Aβ 3-47) [53]. Combining Aβ capture with the N-and C-terminus specific antibodies 6E10 and 4G8 [54], 15 Aβ peptides were detected, including Aβ 1-38 and 1-40 peptides, as well as new peptides truncated in the N-terminus: Aβ 11-40, Aβ 11-42 and Aβ 10-40 [20] (Table 1).

Immuno-precipitation-liquid chromatography-MS (IP-LC-MS/MS) and solid phase extraction-liquid chromatography-tandem MS (SPE-LC-MS/MS)

Tandem MS (MS/MS) is an approach employing two stages of mass analysis in order to selectively fragment a specific ion in a mixture of precursor ions. For proteomics application, MS/MS is usually coupled with a separation method, such as reverse phase liquid chromatography (LC) (see the paragraphs “Mobile phases” and “Analytical columns” below). Performed with a triple quadrupole mass spectrometer, this approach is considered as the gold standard for proteins and peptides quantitation (Table 2). In this context, the most commonly used MS acquisition mode is the selected reaction monitoring (SRM) (Figure 2B). Briefly, peptides are ionized in the electrospray source (ESI) (see the paragraph “Ionisation mode” below). Ions with a given mass (precursor ions) are then selected in the first quadrupole of the mass spectrometer, fragmented in the second quadrupole, and specific ions produced by the fragmentation reaction (product ions) are selected in the third quadrupole for quantification. The precursor ion/product ion couple is called a “transition”.

Table 2:

Comparison of the different mass spectrometry technologies.

Analytical precisionPossible quantificationClinical implementationComplexity of the system and its set-upCost of the system
MALDI+++++++++
SELDI++++
LC-MS++++++++++++++

Recently, candidate reference methods [reference measurement procedure (RMP)] have been proposed for the absolute quantification of Aβ 1-42 by LC-MS/MS [55, 56]. If selected as higher order reference method, they could be used to certify a reference material [certified reference material (CRM)] that would become the “gold standard” with which all in vitro diagnostic kits should be calibrated with to fulfill requirements of the European Commission in vitro diagnostic device directive (IVD Directive 98/79/EC) [57]. This CRM would allow standardizing the different routine methods, thus improving results comparability and reliability.

To set up a SRM method for Aβ peptides, these analytes were initially concentrated by IP (6E10 antibodies) [58]. IP was progressively replaced by solid phase extraction (SPE) which resulted in a very efficient detection of Aβ peptides [40, 42] (Figure 2C). The first SPEs of Aβ peptides used hydrophilic-lipophilic balanced (HLB) [59] and SepPak C18 [60] stationary phases. Recent advances on mixed-mode of SPE with Mild Cation eXchange (MCX), improved the selectivity and recovery of Aβ peptides [40, 42]. Of note, regardless of the prefractionation approach, CSF is generally dried in a lyophilizer or a vacuum centrifuge concentrator (speedvac) in order to concentrate, freeze-dry and store them before analysis. The dry residues are then resolubilized in an alkaline solution (e.g., 20% ACN, 1% NH4OH), in order to avoid aggregation of Aβ peptides before analysis [61].

Mobile phases

The use of reversed phase high pressure LC (HPLC) columns associated with an acid mobile phase is the usual strategy for peptide separation in a wide variety of biological matrices [62, 63]. Quantification of Aβ peptides is however mainly done with an alkaline mobile phase which is more adapted to the physical chemistry of Aβ peptides. As a matter of fact, right from 1992, Burdick et al. showed that aggregation of Aβ peptides increased when pH ranged between 2 and 9 [61]. To avoid aggregation, the chromatographic separation of Aβ 1-40 and 1-42 is therefore better performed under alkaline conditions. Furthermore, the signals obtained in MS under alkaline conditions are about 10-fold higher than those obtained using an acid mobile phase [58]. Several mobile phases have been tested (ammonium acetate, methanol, acetonitrile, hexa-fluoro-isopropanol) and the mix of acetonitrile and ammonium hydroxide was giving the best results in terms of intensity and chromatographic separation of Aβ peptides [58].

Analytical columns

Due to their hydrophobic nature, Aβ peptides can be separated on a C8 [64, 65] or C18-type non-polar column [42, 58]. Separation of Aβ peptides being improved under alkaline conditions, the columns must be resistant to alkaline pH (Table 3). Recently, in order to reduce the contamination of the column by proteins or other hydrophobic molecules, Pannee et al. used a reversed-phase monolith column [42].

Table 3:

Columns used for alkaline mobile phases.

ColumnspH rangeMobile phasesReferences
Waters C8 Xterra MS

(3.0×150 mm, 3.5 μm)
1–12Mobile phase A: 5% ACN 5% Isopropanol 10 mM ammonium acetate

Mobile phase B: 45% ACN 45% Isopropanol 10 mM ammonium acetate
[64]
Waters XBridgeC18 column

(150×2.1 mm id, 3.5 μm, 125 Å)
1–12Mobile phase A: 0.1% NH4OH

Mobile phase B: 0.1% NH4OH ACN (1:200 v/v)
[66]
Waters Acquity UPLC BEH300 C18

(2.1×150 mm, 1.7 μm)
1–12Mobile phase A: 0.3% NH4OH (v/v)

Mobile phase B: 90:10 (v/v) ACN/Mobile phase A
[40]
Thermo Scientific ProSwiftRP-4H11–14Mobile phase A: 0.1% NH4OH 5% ACN

Mobile phase B: 0.03% NH4OH 95% ACN
[42]

Ionization mode

ESI can be done in negative or positive mode, meaning that either negatively charged or positively charged species can be obtained and monitored. In the literature, Aβ peptides are often detected in positive mode but according to Oe et al. [58, 66] sensitivity is improved in the negative mode. In 2011, Lame et al. however demonstrated that working in positive mode, but using alkaline instead of acid mobile phases, led to a significant improvement of the ion signal stability over a 24-h period and a lower detection limit for Aβ peptides.

Quantitative detection of Aβ peptides by LC-MS/MS in a clinical context

For the past 15 years, LC coupled to tandem mass spectrometry (LC-MS/MS) has been increasingly used in clinical laboratories [67], and it has been recently adapted to Aβ peptides detection [64]. Compared to immunological techniques, LC-MS/MS shows a very high specificity, multiplexing capacities and absolute quantification which relies on different standards and calibration strategies as illustrated for Aβ peptides in the next paragraphs.

Use of internal standards

In order to control the variability of the different steps of the protocol (i.e., extraction of Aβ peptides, LC separation, MS ionization and fragmentation), an internal standard can be introduced in the sample at the beginning of the process. Ideally, an internal peptide standard corresponds to a peptide synthetized with the incorporation of stable isotopes such as 13C or 15N. These results in a known mass increment, explaining the term often used of “heavy” standard [68]. To quantify Aβ peptides via LC-MS/MS, the ratio between the MS signal intensity of the endogenous peptide and that of its heavy counterpart present at a known concentration, is generally measured in samples and a set of calibrators [69].

Preparation of Aβ standard solutions

Aβ standards are commercially available in freeze-dried forms rather than in solution to avoid problems linked to the high aggregation propensity of these peptides, especially Aβ 1-42. During their re-suspension, Aβ peptides might however go back to the beta sheet conformation they had prior to freeze-drying and thus could rapidly form new aggregates [70]. To overcome this hurdle, standards are often pretreated with hexafluoroisopropanol (HFIP) [71] in order to denature beta sheets and favor α-helix conformations. Aβ standards need eventually to be re-suspended in solvents compatible with MS analysis, i.e., acetonitrile/ammonium hydroxide (20%:1%, v/v) [42, 66] or dimethylsulfoxide (DMSO) [40].

Calibration curves and model matrices

Several peptide quantification strategies exist (see below and Figure 3). The choice depends of many issues including: the expected values and dynamic range of the analytes, the availability and the cost of light and heavy standards or the nature and the accessibility of a model matrix in which the calibrators will be prepared. Regarding the matrix, it has to be as close as possible as that of the samples to avoid matrix-related effects, and it should not contain any endogenous analytes. For Aβ peptides, it is also important to verify that the model matrix does not induce any aggregation or non-specific adsorption. For quantification of Aβ peptides, several model matrices have been used, including human CSF depleted of Aβ peptides, rat CSF, artificial CSF (Na 150; K 3.0; Ca 1.4; Mg 0.8; P 1.0; Cl 155 in mM, Tocris Bioscience), artificial CSF supplement with 5% of rat plasma or with 4 mg/mL of bovine serum albumin [40, 56, 66, 72].

Figure 3: Different MS-based quantification strategies.(A) The “normal curve” consists in adding a variable quantity of light standard and a constant quantity of heavy standard in a model matrix that mimics the biological fluid of interest. The calibration model is obtained from light standard/heavy standard concentration ratios and light standard/heavy standard peak area ratios. (B) The “reverse curve” is the opposite of the “normal curve”. The light standard is added in a constant quantity in the relevant biological fluid (CSF, model matrix) while the quantity of heavy standard varies. The calibration model is obtained from the areas ratio of the MS signal of the heavy standard over the light standard according to the concentration ratio of the heavy standard over the light standard.
Figure 3:

Different MS-based quantification strategies.

(A) The “normal curve” consists in adding a variable quantity of light standard and a constant quantity of heavy standard in a model matrix that mimics the biological fluid of interest. The calibration model is obtained from light standard/heavy standard concentration ratios and light standard/heavy standard peak area ratios. (B) The “reverse curve” is the opposite of the “normal curve”. The light standard is added in a constant quantity in the relevant biological fluid (CSF, model matrix) while the quantity of heavy standard varies. The calibration model is obtained from the areas ratio of the MS signal of the heavy standard over the light standard according to the concentration ratio of the heavy standard over the light standard.

Normal curve

The “normal curve” approach is commonly used to generate calibration curves (Figure 3A). The procedure is to add to the model matrix a variable amount of light standard and a known amount of heavy standard, which will be also added to each sample. A calibration curve is then established by plotting the ratio between the concentrations of the light and heavy standard as function of the ratio between the peak areas of the transitions used to monitor the light and heavy analytes. The concentration of the endogenous analyte in a clinical sample is then computed from the ratio between the peak areas of the endogenous compound and that of the heavy standard that was added to it.

Reverse curve

On the opposite of the “normal curve”, the procedure is to add a variable amount of the heavy standard to the matrix (which could be a pool of samples) in presence of a known amount of light standard (or endogenous analytes) (Figure 3B). This approach has been sometimes chosen for Aβ peptides since, on one hand, commutability studies suggested that only human CSF was a suitable matrix to mimic clinical samples [55], and on the other hand, it was very difficult to fully deplete CSF of Aβ peptides. The use of this type of calibration curve, with CSF pools as a matrix, may help to more accurately determine limits of detection and quantification of Aβ peptides [42, 73].

Comparison between ELISA and LC-MS/MS

When developing a new analytical strategy, it is important to compare it with the existing ones. Several studies have analyzed samples with both ELISA and LC-MS/MS. In 2006, Oe et al. [66] generated the first results for Aβ peptides quantification after IP and SRM and compared them to those obtained by ELISA. Correlation coefficients obtained were respectively of 0.64 and 0.91 for Aβ 1-40 and Aβ 1-42. In 2013, Pannee et al. [42] developed a MS-based method and obtained lower correlation coefficients ranging from 0.35 to 0.77 and higher Aβ 1-42 concentration values with the MS method. Such discrepancy between ELISA and LC-MS/MS methods are likely related to differences in standard materials (i.e., purity and aggregation level of the light and heavy standards used to prepare the calibrators), as well as differences in the nature of the two methods: while ELISA relies on the immunological recognition of nearby epitopes, LC-MS/MS measures transitions that are generated from peptides which could be located in different regions of the biomarker of interest.

Conclusions

Clinical laboratories measuring Aβ 1-40 and 1-42 CSF levels are mostly using simplex ELISA kits from different companies [4, 14]. Immunodetection approaches are straightforward, but they present some drawbacks such as difficulties to develop good multiplex panels, to detect additional forms of Aβ peptides, or to reach a low variability from batch to batch, and between providers. MS could help resolve these issues with the development of LC-MS/MS reference methods [55, 56] along with the forthcoming availability of a CRM. In addition, MS will be useful to qualify, control and improve comparability of results from existing immunoassays by providing calibrators with target values traceable to a reference method and/or CRM. However, the MS quantification of Aβ peptides will likely stay restricted to expert laboratories since it requires costly mass spectrometers, and highly qualified staff. In addition, the clinical added value of detecting in routine other Aβ peptides than Aβ 1-40 and 1-42 CSF has yet to be demonstrated. Diffusion of the technology in specialized laboratories involved in AD clinical research will anyway benefit from the future development of kits which would include standardized protocols, consumables and reagents (peptide standards).


Corresponding authors: Vincent Delatour, Laboratoire National de Métrologie et d’Essais (LNE), 1 rue Gaston Boissier, Paris, France, E-mail: ; and Sylvain Lehmann, Laboratoire de Biochimie et de Protéomique Clinique, Institut de Médecine Régénérative et Biothérapies (LBPC-IRMB), CHU de Montpellier, 80 rue Augustin Fliche, Montpellier, France, E-mail:

Acknowledgments

This work was in part supported by the “Programme hospitalier de recherche Clinique” (PHRC) “ProMarA: use of targeted quantitative proteomics and metabolic labelling with stable isotopes for the diagnosis and the investigation of neurological disorders and in particular Alzheimer Disease” and the European FP7 Joint Programming on Neurodegenerative Diseases (JPND) BiomarkAPD.

Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

Financial support: None declared.

Employment or leadership: None declared.

Honorarium: None declared.

Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Gulland A. Number of people with dementia will reach 65.7 million by 2030, says report. Brit Med J 2012;344:e2604.10.1136/bmj.e2604Search in Google Scholar

2. Wallon D, Rousseau S, Rovelet-Lecrux A, Quillard-Muraine M, Guyant-Marechal L, Martinaud O, et al. The French series of autosomal dominant early onset Alzheimer’s disease cases: mutation spectrum and cerebrospinal fluid biomarkers. J Alzheimers Dis 2012;30:847–56.10.3233/JAD-2012-120172Search in Google Scholar

3. Petersen RC, Smith GE, Waring SC, Ivnik RJ, Kokmen E, Tangelos EG. Aging, memory, and mild cognitive impairment. Int Psychogeriatr 1997;9(Suppl 1):65–9.10.1017/S1041610297004717Search in Google Scholar

4. Mouton-Liger F, Wallon D, Troussiere AC, Yatimi R, Dumurgier J, Magnin E, et al. Impact of cerebro-spinal fluid biomarkers of Alzheimer’s disease in clinical practice: a multicentric study. J Neurol 2014;261:144–51.10.1007/s00415-013-7160-3Search in Google Scholar

5. Gabelle A, Portet F, Berr C, Touchon J. Neurodegenerative dementia and parkinsonism. J Nutr Health Aging 2010;14:37–44.10.1007/s12603-010-0007-zSearch in Google Scholar

6. Iwatsubo T, Odaka A, Suzuki N, Mizusawa H, Nukina N, Ihara Y. Visualization of a beta 42(43) and a beta 40 in senile plaques with end-specific a beta monoclonals: evidence that an initially deposited species is a beta 42(43). Neuron 1994;13:45–53.10.1016/0896-6273(94)90458-8Search in Google Scholar

7. Grimmer T, Riemenschneider M, Forstl H, Henriksen G, Klunk WE, Mathis CA, et al. Beta amyloid in Alzheimer’s disease: increased deposition in brain is reflected in reduced concentration in cerebrospinal fluid. Biol Psychiatry 2009;65:927–34.10.1016/j.biopsych.2009.01.027Search in Google Scholar

8. Shoji M, Matsubara E, Kanai M, Watanabe M, Nakamura T, Tomidokoro Y, et al. Combination assay of csf tau, a beta 1-40 and a beta 1-42(43) as a biochemical marker of Alzheimer’s disease. J Neurol Sci 1998;158:134–40.10.1016/S0022-510X(98)00122-1Search in Google Scholar

9. Gabelle A, Roche S, Geny C, Touchon J, Lehmann S. Interest of csf markers in positive diagnosis of Alzheimer’s disease in routine clinical practice. Alzheimer Dementia 2009;5:353.10.1016/j.jalz.2009.04.1220Search in Google Scholar

10. DeKosky ST, Carrillo MC, Phelps C, Knopman D, Petersen RC, Frank R, et al. Revision of the criteria for Alzheimer’s disease: a symposium. Alzheimers Dement 2011;7:e1–12.10.1016/j.jalz.2010.12.007Search in Google Scholar PubMed

11. Perrin RJ, Fagan AM, Holtzman DM. Multimodal techniques for diagnosis and prognosis of Alzheimer’s disease. Nature 2009;461:916–22.10.1038/nature08538Search in Google Scholar

12. Dubois B, Feldman HH, Jacova C, Dekosky ST, Barberger-Gateau P, Cummings J, et al. Research criteria for the diagnosis of Alzheimer’s disease: revising the nincds-adrda criteria. Lancet Neurol 2007;6:734–46.10.1016/S1474-4422(07)70178-3Search in Google Scholar

13. Mollenhauer B, Bibl M, Wiltfang J, Steinacker P, Ciesielczyk B, Neubert K, et al. Total tau protein, phosphorylated tau (181p) protein, beta-amyloid(1-42), and beta-amyloid(1-40) in cerebrospinal fluid of patients with dementia with lewy bodies. Clin Chem Lab Med 2006;44:192–5.10.1515/CCLM.2006.035Search in Google Scholar PubMed

14. Gabelle A, Dumurgier J, Vercruysse O, Paquet C, Bombois S, Laplanche JL, et al. Impact of the 2008-2012 French Alzheimer Plan on the use of cerebrospinal fluid biomarkers in research memory center: the plm study. J Alzheimers Dis 2013;34:297–305.10.3233/JAD-121549Search in Google Scholar PubMed

15. Gabelle A, Roche S, Geny C, Bennys K, Labauge P, Tholance Y, et al. Correlations between soluble alpha/beta forms of amyloid precursor protein and abeta38, 40, and 42 in human cerebrospinal fluid. Brain Res 2010;1357:175–83.10.1016/j.brainres.2010.08.022Search in Google Scholar PubMed

16. Borghys H, Jacobs T, Van Broeck B, Dillen L, Dhuyvetter D, Gijsen H, et al. Comparison of two different methods for measurement of amyloid-beta peptides in cerebrospinal fluid after bace1 inhibition in a dog model. J Alzheimers Dis 2014;38:39–48.10.3233/JAD-130599Search in Google Scholar PubMed

17. Olsson A, Vanderstichele H, Andreasen N, De Meyer G, Wallin A, Holmberg B, et al. Simultaneous measurement of beta-amyloid(1-42), total tau, and phosphorylated tau (thr181) in cerebrospinal fluid by the xmap technology. Clin Chem 2005;51:336–45.10.1373/clinchem.2004.039347Search in Google Scholar PubMed

18. Reijn TS, Rikkert MO, van Geel WJ, de Jong D, Verbeek MM. Diagnostic accuracy of elisa and xmap technology for analysis of amyloid beta(42) and tau proteins. Clin Chem 2007;53:859–65.10.1373/clinchem.2006.081679Search in Google Scholar PubMed

19. Shaw LM, Vanderstichele H, Knapik-Czajka M, Clark CM, Aisen PS, Petersen RC, et al. Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann Neurol 2009;65:403–13.10.1002/ana.21610Search in Google Scholar PubMed PubMed Central

20. Maddalena AS, Papassotiropoulos A, Gonzalez-Agosti C, Signorell A, Hegi T, Pasch T, et al. Cerebrospinal fluid profile of amyloid beta peptides in patients with Alzheimer’s disease determined by protein biochip technology. Neurodegener Dis 2004;1:231–5.10.1159/000080991Search in Google Scholar PubMed

21. Perez-Grijalba V, Pesini P, Allue JA, Sarasa L, Montanes M, Lacosta AM, et al. Abeta1-17 is a major amyloid-beta fragment isoform in cerebrospinal fluid and blood with possible diagnostic value in Alzheimer’s disease. J Alzheimers Dis 2015;43:47–56.10.3233/JAD-140156Search in Google Scholar PubMed

22. Jarrett JT, Berger EP, Lansbury PT, Jr. The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer’s disease. Biochemistry 1993;32:4693–7.10.1021/bi00069a001Search in Google Scholar PubMed

23. Fitzpatrick AW, Debelouchina GT, Bayro MJ, Clare DK, Caporini MA, Bajaj VS, et al. Atomic structure and hierarchical assembly of a cross-beta amyloid fibril. Proc Natl Acad Sci USA 2013;110:5468–73.10.1073/pnas.1219476110Search in Google Scholar PubMed PubMed Central

24. Slemmon JR, Meredith J, Guss V, Andreasson U, Andreasen N, Zetterberg H, et al. Measurement of abeta1-42 in cerebrospinal fluid is influenced by matrix effects. J Neurochem 2012;120:325–33.10.1111/j.1471-4159.2011.07553.xSearch in Google Scholar PubMed

25. Perret-Liaudet A, Pelpel M, Tholance Y, Dumont B, Vanderstichele H, Zorzi W, et al. Cerebrospinal fluid collection tubes: a critical issue for Alzheimer disease diagnosis. Clin Chem 2012;58:787–9.10.1373/clinchem.2011.178368Search in Google Scholar PubMed

26. Del Campo M, Mollenhauer B, Bertolotto A, Engelborghs S, Hampel H, Simonsen AH, et al. Recommendations to standardize preanalytical confounding factors in Alzheimer’s and parkinson’s disease cerebrospinal fluid biomarkers: an update. Biomark Med 2012;6:419–30.10.2217/bmm.12.46Search in Google Scholar PubMed

27. Vanderstichele H, Bibl M, Engelborghs S, Le Bastard N, Lewczuk P, Molinuevo JL, et al. Standardization of preanalytical aspects of cerebrospinal fluid biomarker testing for Alzheimer’s disease diagnosis: a consensus paper from the Alzheimer’s biomarkers standardization initiative. Alzheimers Dement 2012;8:65–73.10.1016/j.jalz.2011.07.004Search in Google Scholar PubMed

28. Mattsson N, Andreasson U, Persson S, Carrillo MC, Collins S, Chalbot S, et al. Csf biomarker variability in the Alzheimer’s Association quality control program. Alzheimers Dement 2013;9:251–61.10.1016/j.jalz.2013.01.010Search in Google Scholar PubMed PubMed Central

29. Poceta JS, Parsons L, Engelland S, Kripke DF. Circadian rhythm of csf monoamines and hypocretin-1 in restless legs syndrome and parkinson’s disease. Sleep Med 2009;10:129–33.10.1016/j.sleep.2007.11.002Search in Google Scholar PubMed

30. Bateman RJ, Wen G, Morris JC, Holtzman DM. Fluctuations of csf amyloid-beta levels: Implications for a diagnostic and therapeutic biomarker. Neurology 2007;68:666–9.10.1212/01.wnl.0000256043.50901.e3Search in Google Scholar PubMed

31. Bjerke M, Portelius E, Minthon L, Wallin A, Anckarsater H, Anckarsater R, et al. Confounding factors influencing amyloid beta concentration in cerebrospinal fluid. Int J Alzheimers Dis 2010;2010.10.4061/2010/986310Search in Google Scholar PubMed PubMed Central

32. Lewczuk P, Beck G, Esselmann H, Bruckmoser R, Zimmermann R, Fiszer M, et al. Effect of sample collection tubes on cerebrospinal fluid concentrations of tau proteins and amyloid beta peptides. Clin Chem 2006;52:332–4.10.1373/clinchem.2005.058776Search in Google Scholar PubMed

33. Lehmann S, Schraen S, Quadrio I, Paquet C, Bombois S, Delaby C, et al. Impact of harmonization of collection tubes on Alzheimer’s disease diagnosis. Alzheimers Dement 2014;10(5 Suppl):S390–4.e2.10.1016/j.jalz.2013.06.008Search in Google Scholar PubMed

34. Perret-Liaudet A, Pelpel M, Tholance Y, Dumont B, Vanderstichele H, Zorzi W, et al. Risk of Alzheimer’s disease biological misdiagnosis linked to cerebrospinal collection tubes. J Alzheimers Dis 2012;31:13–20.10.3233/JAD-2012-120361Search in Google Scholar PubMed

35. Schoonenboom NS, Mulder C, Vanderstichele H, Van Elk EJ, Kok A, Van Kamp GJ, et al. Effects of processing and storage conditions on amyloid beta (1-42) and tau concentrations in cerebrospinal fluid: implications for use in clinical practice. Clin Chem 2005;51:189–95.10.1373/clinchem.2004.039735Search in Google Scholar PubMed

36. Teunissen CE, Petzold A, Bennett JL, Berven FS, Brundin L, Comabella M, et al. A consensus protocol for the standardization of cerebrospinal fluid collection and biobanking. Neurology 2009;73:1914–22.10.1212/WNL.0b013e3181c47cc2Search in Google Scholar PubMed PubMed Central

37. Simonsen AH, Bahl JM, Danborg PB, Lindstrom V, Larsen SO, Grubb A, et al. Pre-analytical factors influencing the stability of cerebrospinal fluid proteins. J Neurosci Meth 2013;215:234–40.10.1016/j.jneumeth.2013.03.011Search in Google Scholar PubMed

38. Gabelle A, Roche S, Lehmann S. Csf biomarkers: proteomics investigations and clinical applications in neurodegenerative disorders. Rev Neurol (Paris) 2009;165:213–22.10.1016/j.neurol.2008.05.004Search in Google Scholar PubMed

39. Portelius E, Zetterberg H, Gobom J, Andreasson U, Blennow K. Targeted proteomics in Alzheimer’s disease: focus on amyloid-beta. Expert Rev Proteomics 2008;5:225–37.10.1586/14789450.5.2.225Search in Google Scholar PubMed

40. Lame ME, Chambers EE, Blatnik M. Quantitation of amyloid beta peptides abeta(1-38), abeta(1-40), and abeta(1-42) in human cerebrospinal fluid by ultra-performance liquid chromatography-tandem mass spectrometry. Anal Biochem 2011;419:133–9.10.1016/j.ab.2011.08.010Search in Google Scholar PubMed

41. Portelius E, Westman-Brinkmalm A, Zetterberg H, Blennow K. Determination of beta-amyloid peptide signatures in cerebrospinal fluid using immunoprecipitation-mass spectrometry. J Proteome Res 2006;5:1010–6.10.1021/pr050475vSearch in Google Scholar PubMed

42. Pannee J, Portelius E, Oppermann M, Atkins A, Hornshaw M, Zegers I, et al. 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 2013;33:1021–32.10.3233/JAD-2012-121471Search in Google Scholar PubMed

43. Delaby C, Gabelle A, Meynier P, Loubiere V, Vialaret J, Tiers L, et al. Development and validation of dried matrix spot sampling for the quantitative determination of amyloid beta peptides in cerebrospinal fluid. Clin Chem Lab Med 2014;52:649–55.10.1515/cclm-2013-0611Search in Google Scholar PubMed

44. Portelius E, Zetterberg H, Andreasson U, Brinkmalm G, Andreasen N, Wallin A, et al. An Alzheimer’s disease-specific beta-amyloid fragment signature in cerebrospinal fluid. Neurosci Lett 2006;409:215–9.10.1016/j.neulet.2006.09.044Search in Google Scholar PubMed

45. Verpillot R, Esselmann H, Mohamadi MR, Klafki H, Poirier F, Lehnert S, et al. Analysis of amyloid-beta peptides in cerebrospinal fluid samples by capillary electrophoresis coupled with lif detection. Anal Chem 2011;83:1696–703.10.1021/ac102828fSearch in Google Scholar PubMed

46. Portelius E, Tran AJ, Andreasson U, Persson R, Brinkmalm G, Zetterberg H, et al. Characterization of amyloid beta peptides in cerebrospinal fluid by an automated immunoprecipitation procedure followed by mass spectrometry. J Proteome Res 2007;6:4433–9.10.1021/pr0703627Search in Google Scholar PubMed

47. Portelius E, Dean RA, Gustavsson MK, Andreasson U, Zetterberg H, Siemers E, et al. A novel abeta isoform pattern in csf reflects gamma-secretase inhibition in Alzheimer disease. Alzheimers Res Ther 2010;2:7.10.1186/alzrt30Search in Google Scholar PubMed PubMed Central

48. Portelius E, Mattsson N, Andreasson U, Blennow K, Zetterberg H. Novel abeta isoforms in Alzheimer’s disease – their role in diagnosis and treatment. Curr Pharm Des 2011;17:2594–602.10.2174/138161211797416039Search in Google Scholar PubMed

49. Portelius E, Brinkmalm G, Tran A, Andreasson U, Zetterberg H, Westman-Brinkmalm A, et al. Identification of novel n-terminal fragments of amyloid precursor protein in cerebrospinal fluid. Exp Neurol 2010;223:351–8.10.1016/j.expneurol.2009.06.011Search in Google Scholar PubMed

50. Portelius E, Gustavsson MK, Zetterberg H, Andreasson U, Blennow K. Evaluation of the performance of novel abeta isoforms as theragnostic markers in Alzheimer’s disease: from the cell to the patient. Neurodegener Dis 2012;10:138–40.10.1159/000334537Search in Google Scholar PubMed

51. Nutu M, Bourgeois P, Zetterberg H, Portelius E, Andreasson U, Parent S, et al. Abeta1-15/16 as a potential diagnostic marker in neurodegenerative diseases. Neuromolecular Med 2013;15:169–79.10.1007/s12017-012-8208-8Search in Google Scholar

52. Zetterberg H, Ruetschi U, Portelius E, Brinkmalm G, Andreasson U, Blennow K, et al. Clinical proteomics in neurodegenerative disorders. Acta Neurol Scand 2008;118:1–11.10.1111/j.1600-0404.2007.00985.xSearch in Google Scholar

53. Lewczuk P, Esselmann H, Groemer TW, Bibl M, Maler JM, Steinacker P, et al. Amyloid beta peptides in cerebrospinal fluid as profiled with surface enhancement laser desoprtion/ionization time of flight mass spectrometry: evidence of novel biomarkers in Alzheimer’s disease Biol Psychiatry 2003;55: 524–30.10.1016/j.biopsych.2003.10.014Search in Google Scholar

54. Albertini V, Bruno A, Paterlini A, Lista S, Benussi L, Cereda C, et al. Optimization protocol for amyloid-beta peptides detection in human cerebrospinal fluid using seldi tof ms. Proteomics Clin Appl 2010;4:352–7.10.1002/prca.200900166Search in Google Scholar

55. Leinenbach A, Pannee J, Dulffer T, Huber A, Bittner T, Andreasson U, et al. Mass spectrometry-based candidate reference measurement procedure for quantification of amyloid-beta in cerebrospinal fluid. Clin Chem 2014;60:987–94.10.1373/clinchem.2013.220392Search in Google Scholar

56. Korecka M, Waligorska T, Figurski M, Toledo JB, Arnold SE, Grossman M, et al. 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 2014;41:441–51.10.3233/JAD-132489Search in Google Scholar

57. Directive 98/79/EC of the European Parliament and of the Council of 27 October 1998 on In Vitro Diagnostic Medical Devices OJECL-L.Search in Google Scholar

58. Schneider B, Pietri M, Mouillet-Richard S, Ermonval M, Mutel V, Launay JM, et al. Control of bioamine metabolism by 5-ht2b and alpha 1d autoreceptors through reactive oxygen species and tumor necrosis factor-alpha signaling in neuronal cells. Ann NY Acad Sci 2006;1091:123–41.10.1196/annals.1378.061Search in Google Scholar

59. Lanz TA, Schachter JB. Solid-phase extraction enhances detection of beta-amyloid peptides in plasma and enables abeta quantification following passive immunization with abeta antibodies. J Neurosci Meth 2008;169:16–22.10.1016/j.jneumeth.2007.11.009Search in Google Scholar

60. Ditto NT, Kline TR, Alfinito PD, Slemmon JR. Enrichment and analysis of Alzheimer’s abeta1-42 peptide in human plasma and whole blood. J Neurosci Meth 2009;182:260–5.10.1016/j.jneumeth.2009.05.025Search in Google Scholar

61. Burdick D, Soreghan B, Kwon M, Kosmoski J, Knauer M, Henschen A, et al. Assembly and aggregation properties of synthetic Alzheimer’s a4/beta amyloid peptide analogs. J Biol Chem 1992;267:546–54.10.1016/S0021-9258(18)48529-8Search in Google Scholar

62. Link AJ, Eng J, Schieltz DM, Carmack E, Mize GJ, Morris DR, et al. Direct analysis of protein complexes using mass spectrometry. Nat Biotechnol 1999;17:676–82.10.1038/10890Search in Google Scholar

63. Gygi SP, Rist B, Gerber SA, Turecek F, Gelb MH, Aebersold R. Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nat Biotechnol 1999;17:994–9.10.1038/13690Search in Google Scholar

64. Du C, Ramaley C, McLean H, Leonard SC, Miller J. High-performance liquid chromatography coupled with tandem mass spectrometry for the detection of amyloid beta peptide related with Alzheimer’s disease. J Biomol Tech 2005; 16:356–63.Search in Google Scholar

65. Thompson AJ, Lim TK, Barrow CJ. On-line high-performance liquid chromatography/mass spectrometric investigation of amyloid-beta peptide variants found in Alzheimer’s disease. Rapid Commun Mass Sp 1999;13:2348–51.10.1002/(SICI)1097-0231(19991215)13:23<2348::AID-RCM797>3.0.CO;2-JSearch in Google Scholar

66. Oe T, Ackermann BL, Inoue K, Berna MJ, Garner CO, Gelfanova V, et al. Quantitative analysis of amyloid beta peptides in cerebrospinal fluid of Alzheimer’s disease patients by immunoaffinity purification and stable isotope dilution liquid chromatography/negative electrospray ionization tandem mass spectrometry. Rapid Commun Mass Sp 2006;20:3723–35.10.1002/rcm.2787Search in Google Scholar

67. Grebe SK, Singh RJ. Lc-ms/ms in the clinical laboratory – where to from here? Clin Biochem Rev 2011;32:5–31.Search in Google Scholar

68. Lehmann S, Hoofnagle A, Hochstrasser D, Brede C, Glueckmann M, Cocho JA, et al. Quantitative clinical chemistry proteomics (qccp) using mass spectrometry: General characteristics and application. Clin Chem Lab Med 2013;51:919–35.10.1515/cclm-2012-0723Search in Google Scholar

69. Henrion A. Reduction of systematic-errors in quantitative-analysis by isotope-dilution mass-spectrometry (idms) – an iterative method. Fresen J Anal Chem 1994;350:657–8.10.1007/BF00323658Search in Google Scholar

70. Stine WB Jr., Dahlgren KN, Krafft GA, LaDu MJ. In vitro characterization of conditions for amyloid-beta peptide oligomerization and fibrillogenesis. J Biol Chem 2003;278:11612–22.10.1074/jbc.M210207200Search in Google Scholar

71. Ryan TM, Caine J, Mertens HD, Kirby N, Nigro J, Breheney K, et al. Ammonium hydroxide treatment of abeta produces an aggregate free solution suitable for biophysical and cell culture characterization. Peer J 2013;1:e73.10.7717/peerj.73Search in Google Scholar

72. Dillen L, Cools W, Vereyken L, Timmerman P. A screening uhplc-ms/ms method for the analysis of amyloid peptides in cerebrospinal fluid of preclinical species. Bioanalysis 2011;3:45–55.10.4155/bio.10.163Search in Google Scholar

73. Campbell J, Rezai T, Prakash A, Krastins B, Dayon L, Ward M, et al. Evaluation of absolute peptide quantitation strategies using selected reaction monitoring. Proteomics 2011;11:1148–52.10.1002/pmic.201000511Search in Google Scholar

Received: 2014-10-25
Accepted: 2015-1-20
Published Online: 2015-2-18
Published in Print: 2015-9-1

©2015 by De Gruyter

Downloaded on 24.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2014-1048/html
Scroll to top button