Skip to main content
Erschienen in: Neurology and Therapy 2/2019

Open Access 01.12.2019 | Review

A Review on MS-Based Blood Biomarkers for Alzheimer’s Disease

verfasst von: Patrick Oeckl, Markus Otto

Erschienen in: Neurology and Therapy | Sonderheft 2/2019

Abstract

Alzheimer’s disease (AD) is the most common form of neurodegenerative dementia and there is no cure to date. Biomarkers in cerebrospinal fluid (CSF) are already included in the diagnostic work-up of symptomatic patients but markers for preclinical diagnosis and disease progression are not available. Furthermore, blood biomarkers are highly appreciated because they are minimally invasive and more accessible in primary care and in clinical studies. Mass spectrometry (MS) is an established tool for the measurement of various analytes in biological fluids such as blood. Its major strength is the high selectivity which is why it is also preferred as a reference method for immunoassays. MS has been used in several studies in the past for blood biomarker discovery and validation in AD using targeted MS such as multiple/selected reaction monitoring (MRM/SRM) or unbiased approaches (proteomics, metabolomics). In this short review, we give an overview on the status of current MS-based biomarker candidates for AD in blood plasma and serum.
Plain Language Summary: Plain language summary available for this article.
Hinweise

Enhanced digital features

To view enhanced digital features for this article go to https://​doi.​org/​10.​6084/​m9.​figshare.​9792635.
Key Summary Points
Mass spectrometry (MS) is an established tool to measure analytes in blood.
Reliable MS-based biomarkers in blood for Alzheimer´s disease (AD) are still rare.
Most promising MS-based biomarkers for AD at the moment seem to be the determination of Aβ peptides by IP-MS and the “genotyping” of ApoE by MRM.
MS will be essential in the definition of reference methods for the measurement of AD biomarkers in blood.

Plain Language Summary

Alzheimer´s disease (AD) is the most common form of neurodegenerative dementia and there is no cure to date. A biomarker is a naturally occurring molecule, gene, or characteristic by which a particular disease can be identified or its progression monitored. Biomarkers in cerebrospinal fluid are used in clinical diagnosis of AD, but markers for pre-clinical diagnosis and disease progression are not available. Blood biomarkers are accessible in primary care in a minimally invasive way, convenient to patients, with little cost and suitable for repeated sampling for longitudinal assessments. Mass spectrometry (MS) is an established analytical tool for measuring markers in biological fluids such as the blood, with the benefit of higher selectivity than alternative methods. MS has been used in several studies in the past for blood biomarker discovery and validation in AD and a review of the available research identified several biomarker candidates, of which the most promising are Aβ peptides, proteins released by AD pathology, and ApoE genotyping, although both markers require targeted, specific methods of MS and require further research for validation. Mass spectrometry is expected to be an essential tool in the definition of reference methods for the measurement of AD biomarkers in blood.

Introduction

Alzheimer´s disease (AD) is a neurodegenerative disease and the most common form of dementia. The characteristic feature of AD is the formation of amyloid plaques and neurofibrillary tangles in the brain accompanied with the degeneration of synapses. To date, the cause of AD is still unclear and there is no cure available making it a serious health problem in the aging societies of industrial countries [1]. Diagnosis is mainly based on clinical symptoms, although the measurement of tau protein and the amyloid β peptide (Aβ42) in cerebrospinal fluid (CSF) was included into the diagnostic criteria in 2011 to support AD neuropathology [2]. Biomarkers for presymptomatic diagnosis and disease progression are not yet available but are essential for early treatment of patients and drug development.
In contrast to CSF, the measurement of biomarkers in blood has several advantages: it is more convenient for patients, it is less costly and it is more suitable for repeated sample collection in longitudinal assessments. Mass spectrometry (MS) is a valuable tool for biomarker measurement in blood (reviewed in [3]). Its major strength is the high selectivity. This is an important advantage compared with immunoassays which often struggle with matrix effects and unspecific signals in blood samples. In MS, the target analyte is measured by its mass-to-charge ratio (m/z). In modern hybrid MS instruments more than one mass analyzer is included. This allows additional fragmentation of the target analyte and quantification of the specific fragments. This approach is called multiple/selected reaction monitoring (MRM/SRM) or parallel reaction monitoring (PRM) depending on the mass analyzer used. It significantly increases specificity compared with single m/z measurement. MS can also be used for unbiased discovery of new biomarker candidates (e.g. proteomics, metabolomics). Here, a successive acquisition of full-scan and fragment mass spectra is performed followed by matching with a database to identify the measured analytes. Very often, MS is coupled to gas or liquid chromatography to perform an additional separation of molecules before MS analysis.
In this short review we give an overview on biomarker (candidates) for AD in blood plasma and serum which were measured using MS techniques.

Methods

We searched the PubMed database in June 2019 for articles matching the following search terms: Alzheimer’s disease AND blood or plasma or serum AND mass spectrometry or MRM or SRM or proteomics or proteomic or proteome or metabolomics or metabolomic or metabolome or lipidomics or lipidomic or lipidome or glycomics or glycomic or glycome. A total of 483 articles were found. After exclusion of review articles, the titles and abstracts of 422 articles were screened for studies using MS-based techniques for measuring endogenous molecules in serum/plasma (not blood cells) from human AD patients. From the remaining 151 articles, we further excluded articles which used MS for protein identification only (not quantification). In order to focus on the most reliable biomarker candidates, we included unbiased discovery approaches only if the identified candidates were verified using MRM/PRM (proteomics) or using synthetic compounds (metabolomics). With the exception of the Mapstone panel (examined in several studies), we also did not include biomarker panels because they were unique to the respective study populations. Finally, 66 articles fulfilled our criteria.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.

Results

Aβ Peptides

The accumulation of Aβ in amyloid plaques in the brain is a major hallmark of AD. Alterations of the proteolytic APP (amyloid precursor protein) processing and release of the Aβ peptides is a key event in AD pathophysiology [1]. The measurement of Aβ42 in CSF is an established AD biomarker of amyloid pathology and included in the diagnostic criteria [2]. MRM is used as a reference method for Aβ measurement in CSF [4, 5]. Pannee and colleagues used their antibody-free CSF reference MRM also to investigate blood levels of Aβ38, Aβ40 and Aβ42. In their small cohort of 9 AD and 10 control patients, they did not observe significant differences between the two groups [6]. There was also no correlation of Aβ42 levels in plasma and CSF but CSF was analyzed by ELISA. They also characterized additional Aβ variants in plasma by immunoprecipitation (IP) and MALDI-ToF MS (Aβ15, Aβ17, Aβ19, Aβ20, Aβ33, Aβ34, Aβ5-40, Aβ37, Aβ3-40 and Aβ39) but observed no differences between AD and control samples (n = 2).
Another group also reported an antibody-free MRM method with the aim to measure total Aβ in plasma [7]. However, they performed tryptic digestion of all plasma proteins as an initial step of sample preparation. The tryptic peptide (Aβ17-28) selected for Aβ quantification in this study is not specific for Aβ and is also released from full-length APP during tryptic digestion. Thus, measurement results did not represent Aβ in plasma. This is also reflected in the reported concentration which is more than tenfold higher compared with the more reliable method from Pannee et al. [6].
A Japanese group published a first study in 2014 using IP-MALDI ToF MS to measure Aβ peptide levels in plasma. They investigated subjects with and without cerebral amyloid deposition which was determined by positron emission tomography (PET) with Pittsburgh Compound B (PiB) [8]. The IP-MALDI ToF MS method used was already validated in terms of precision and dilution linearity previously [9]. The authors chose this pathology-driven approach because AD-like amyloid deposition is also found in a substantial number of non-demented aged individuals [10]. In this context, a syndromal comparison (i.e. AD patients vs controls) might underestimate the diagnostic performance of amyloid-related plasma biomarkers. Thereby, they also followed the recent suggestion of a biological definition of AD by the ATN classification of biomarkers [11]. Kaneko et al. measured plasma levels of Aβ40, Aβ42 and APP669-711 in 22 PiB-negative and 40 PiB-positive (including AD, MCI and healthy controls) subjects. They observed significantly reduced Aβ42 and Aβ42/Aβ40 and increased APP669-711/Aβ42 in PiB-positive vs PiB negative subjects [8]. Subgroup comparison showed, that the APP669-711/Aβ42 ratio was also increased in PiB-positive AD compared with PiB-positive healthy controls. The ratio APP669-711/Aβ42 yielded a high sensitivity and specificity (93% and 96%) for the detection of cerebral amyloid pathology. The authors confirmed their results recently with a slightly modified method in two additional patient cohorts from the Japanese National Center for Geriatrics and Gerontology (NCGG, n = 121) and Australian Imaging, Biomarker and Lifestyle Study of Ageing (AIBL, n = 252) [12]. APP669-711/Aβ42 and Aβ40/Aβ42 showed good discriminatory power for amyloid positive and negative subjects in the NCGG cohort (sensitivity 68% and 96%, specificity 92% and 87%) and AIBL cohort (sensitivity 83% and 85%, specificity 70% and 68%). In addition, they created a composite biomarker from both ratios with even better performance in the NCGG and AIBL cohort (sensitivity 86% and 86%, specificity 89% and 78%). They used stable isotope-labeled (SIL)-Aβ38 as internal standard (IS) for all Aβ peptides, which has the advantage that the Aβ ratios are independent from the spiked amount of IS. Therefore, the established cut-off values are transferable. A drawback could be that the different properties of the Aβ peptides are not optimally reflected by Aβ38. This might contribute to the center differences of the Aβ ratios between the NCGG and AIBL cohort due to different matrix effects.
Most recently, the Bateman group published a study confirming the good predictive power of Aβ40/Aβ42 in plasma for amyloid PET status [13]. They used tryptic digestion and MRM instead of MALDI-ToF but observed a similar sensitivity (88%) and specificity (76%) for the discrimination of amyloid positive and negative subjects compared with Nakamura et al. [12].The high performance for detection of cerebral amyloid pathology and the consistent findings with IP-MS methods in multiple cohorts and by different research groups makes it a very promising MS-based biomarker.

Phospholipids/Sphingolipids

Phospholipids are an integral part of the plasma membrane and their composition substantially influences the function of membrane proteins, receptors, enzymes and ion channels. The reduced phospholipid amount in AD brains gives a first link of phospholipids to AD pathophysiology. Furthermore, the cleavage of APP, releasing the Aβ peptides, takes place at the cell membrane and thus in direct contact with phospholipids [14].
In 2014, Mapstone and colleagues [15] reported a panel of phospholipids (Table 1) with significant changes in plasma already in the preclinical phase of AD. The phospholipids and other metabolites were measured using a commercially available MRM kit. They compared AD/amnestic MCI (mild cognitive impairment) patients with cognitively normal controls from a longitudinal observational study. They especially looked at subjects converting from cognitively normal to AD/aMCI during the observational period. Here, they included plasma samples from the time before and after conversion. Combining the panel of eight phospholipids with two acylcarnitines (C3 and C16:1-OH, Table 1), they were able to identify AD/aMCI patients in their cohort of 106 patients at a time, when they were cognitively normal (on average 2.1 years before conversion). The sensitivity and specificity was 90%. They successfully validated their results in a different subset of 40 patients from the same cohort. Significant changes in AD, MCI or dementia of four lipids from the Mapstone panel (PC aa C36:6, PC aa C38:6, LysoPC 18:2 and C3) were also reported in other studies [1621] (see Table 1) but sometimes in the opposite direction [16].
Table 1
Panel of ten lipid biomarkers suggested by Mapstone et al. [15] for detection of preclinical AD
Biomarker
Observation of significant changes in studies
Phospholipids
 
PC aa C36:6
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15]
OR Dem vs Con < 1 [17]
PC aa C38:0
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15]
PC aa C38:6
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15], OR AD < 1 [21]
PC aa C40:1
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15]
PC aa C40:2
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15]
PC aa C40:6
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15]
PC ae C40:6
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15]
LysoPC a C18:2
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15], AD vs Con↓ [19], OR MCI vs Con > 1 [16], OR MCI/Dem vs Con > 1 [16], Concentration increase during follow-up in AD-converters vs Con↑ [18]
Acylcarnitines
 
C3
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15], MCI vs Con↓ [20]
C16:1-OH
Converter(Pre) vs Con↓ [15], aMCI/AD vs Con↓ [15]
AD Alzheimer’s disease, aMCI amnestic MCI, Con control patients, Dem dementia, MCI mild cognitive impairment, OR odds ratio, Pre samples before conversion to aMCI/AD
↑ Significantly increased, ↓ significantly decreased
Three studies from two other research groups evaluated the Mapstone panel of blood markers in different longitudinal cohorts. These included subjects converting to AD or MCI during the follow-up and the use of the same MRM kit [16, 17, 22]. Casanova et al. investigated samples from two large cohorts of patients, the Baltimore Longitudinal Study of Aging (BLSA) and the Age, Gene/Environment Susceptibility-Reykjavik Study (AGES-RS) comprising 392 patients in total [22]. They could not reproduce the high predictive performance of the Mapstone panel in blood for preclinical AD. In their much larger cohort of preclinical AD converters (n = 93 in BLSA cohort, n = 100 in AGES-RS cohort vs. n = 28 in Mapstone cohort), they observed sensitivities and specificities of 52% and 66% (BLSA cohort) and 47% and 36% (AGES-RS cohort). The use of different sample matrices (plasma vs serum) has been discussed as a possible reason for the discrepancy between these two studies. However, Casanova et al. also provided data that plasma and serum levels of the Mapstone panel are highly correlated [22].
Li and colleagues investigated plasma samples from the Atherosclerosis Risk in Communities Neurocognitive Study (ARIC-NCS). A large majority of these samples were from African Americans (> 96%) and their aim was to evaluate the predictive performance of the Mapstone panel (established in a white population) in African American [16]. Their cohort included 95 MCI/dementia converters with a mean follow-up time of 7.3 years, which is considerably longer than in the Mapstone study (2.1 years). In agreement with the study by Casanova et al., they observed neither a large added value of the Mapstone panel for prediction of MCI/dementia [16] nor for detection of MCI/dementia in a cross-sectional comparison (n = 441) [17]. However, the MCI/dementia patients in the studies by Li et al. [16, 17] are not solely AD patients which might weaken the discriminatory performance of the (potentially AD-specific) biomarker panel. Changes of several other phospholipids in serum/plasma were reported in single studies and are listed in Table 2.
Table 2
Summary of molecules measured by MS techniques and with a reported change in AD
Molecule
Observed changes
Molecule
Observed changes
Acyl carnitines
 
Cholesterol metabolism
 
Carnitine
MCI vs Con↓ [20], AD vs Con↓ [35]
24S-OH cholesterol esters
AD vs Con↓ [53], MCInon-conv vs MCIconv↑ [53]
 
AD vs MCI↓ [35]
 
MCI vs AD↑ [53]
C5-OH
AD vs Con↓ [20]
27-OH cholesterol
AD vs Con↓ [41]↔[42]
C10:0
AD vs Con↑ [36]↓ [20, 35]
 
MCI vs Con↓ [41]↔[42]
 
MCI vs Con↑ [36]↓ [20]
Desmosterol
AD vs Con ↓ [54], MCI vs Con↓ [54]
 
AD vs MCI↓ [35]
  
C14:2
AD vs Con↓ [35], AD vs MCI↓ [35]
Steroids
 
C10:1
AD vs Con↓ [36], MCI vs Con↓ [36]
5α-Androstane-3α,17β-diol sulfate/5α-Androstane-3α,17β-diol
AD vs Con↓ [55]
 
MCI vs AD↑ [35]
5α-Androstane-3β,17β-diol sulfate/5α-Androstane-3β,17β-diol
AD vs Con↓ [55]
C16:1
AD vs Con↓ [20], MCI vs Con↓ [20]
Free 7α-Hydroxy-DHEA
AD vs Con↑ [56]
C6:1
AD vs Con↓ [20]
7α-Hydroxy-DHEA-S
AD vs Con↑ [56]
C18:2
AD vs Con↓ [20]
7α-Hydroxy-DHEA-FA esters
AD vs Con↑ [56]
C3-DC
AD vs Con↓ [20]
Androsterone sulfate/androsterone
AD vs Con↓ [55]
C14
AD vs Con↓ [20]
Cortisol
OR AD vs Con↑ [57]
C16
AD vs MCI↓ [20]
DHEA-S
AD vs Con↓ [58]
C18:1
AD vs Con↓ [20], MCI vs Con↓ [20]
DHEA-S/DHEA
AD vs Con↓ [55]
 
AD vs MCI↓ [35]
Epiandrosterone sulfate/epiandrosterone
AD vs Con↓ [55]
C18
AD vs Con↓ [20], MCI vs Con↓ [20]
  
  
Ox. stress-med. DHEA-increase
AD vs Con↓ [59], MCI vs Con↑ [59], MCI vs AD↑ [59]
Fatty acids/DAG/TG
   
C14:0
AD vs Con↓ [46]
Vitamin D
 
C16:0
AD vs Con↓ [46]
25(OH) Vitamin D
HR AD with low levels↑ [60]↔[61]
C16:1
AD vs Con↓ [46]
  
C18:0
AD vs Con↓ [46]
Amino acids and derivatives
 
C18:1
AD vs Con↓ [46]
Aspartate
 AD vs Con↓ [36], MCI vs Con↓ [36]
C18:2
AD vs Con↓ [46]
Carnosine
 AD vs Con↓ [62]
γ-C18:3
AD vs Con↓ [46]
Cysteine
MCI vs Con↑ [63]
C18:3
AD vs Con↑ [46]
DOPA
AD vs Con↑ [62]
C20:2
AD vs Con↓ [46]
Glutamic acid
HR AD↑ [64]
C20:5
AD vs Con↓ [47]
Hcy-Cys
AD vs Con↑ [36]
C22:6
AD vs Con↓ [46]
Histidine
AD vs Con↓ [36], AD vs MCI↓ [36]
MMA
AD vs Con [48]
Isoaspartate peptides
AD/MCI vs Con↑ [65]
DAG (34:3)
MCI vs Con↑ [49], Dem vs Con↑ [49]
Methionine
AD vs Con↓ [36], AD vs MCI↓ [36]
DAG (36:3)
MCI vs Con↑ [49], Dem vs Con↑ [49]
Methyldopa
AD vs Con [48]
DAG (36:4)
MCI vs Con↑ [49], Dem vs Con↑ [49]
N-acetylglutamine
AD vs Con↓ [19]
DAG (38:6)
MCI vs Con↑ [49], Dem vs Con↑ [49]
Phe Phe
AD vs Con↑ [36], MCI vs Con↑ [36]
DAG (34:2)
MCI vs Con↑ [49], Dem vs Con↑ [49]
O-Acetylserine
AD vs Con [48]
DAG (36:2)
MCI vs Con↑ [49], Dem vs Con↑ [49]
Tyrosine
AD vs Con [48]
DAG (38:4)
MCI vs Con↑ [49], Dem vs Con↑ [49]
Valine
AD vs Con [48]
TG (48:0)
AD vs Con [48]
  
TG (50:4)
AD vs Con [48]
Metals
 
TG (48:2)
AD vs Con [48]
Al
 AD vs Con↓ [44]↑ [66], MCI vs Con↑ [66]
TG (51:3)
AD vs Con [48]
Al-Ferritin
 AD vs Con↑ [67]
TG (54:6)
AD vs Con [48]
Ca
DLB vs AD↑ [68]
TG (50:3)
AD vs Con [48]
Cd
severeAD vs mildAD↑ [69], AD vs Con↑ [44]
TG (48:1)
AD vs Con [48]
Cs
AD vs Con↓ [45]
TG (52:4)
AD vs Con [48]
Cu
DLB vs AD↑ [68]
TG (48:3)
AD vs Con [48]
Non-ceruloplasmin Cu
MCI vs Con↓ [70], AD vs Con↓ [70]
TG (46:2)
AD vs Con [48]
Fe
AD vs Con↓ [71]
TG (52:5)
AD vs Con [48]
Transferrin-Fe/Transferrin
AD vs Con↓ [71]
TG (58:10)
AD vs Con [48]
Mg
DLB vs AD↑ [68]
TG (56:7)
AD vs Con [48]
Mn
severeAD vs mildAD↓ [69]
TG (56:8)
AD vs Con [48]
 
AD vs Con↑ [45]↓ [66, 72], MCI vs Con↓ [66]
  
Mo
severeAD vs mildAD↑ [69]
Phospholipids/Sphingolipids
 
Se
AD vs Con↓ [45], MCI vs AD↑ [66]
Lyso-PAF (C16:0)
ADconv vs Con↑ [18]
Sn
AD vs Con↑ [44]
Lyso-PAF (C18:0)
ADconv vs Con↑ [18]
Zn
DLB vs AD↓ [68], AD vs Con↓ [66]
Lyso-PAF (C18:1)
ADconv vs Con↑ [18]
  
LysoPC (16:0)
ADconv vs Con↑ [18]
Oxidative stress markers
 
LysoPC (18:0)
ADconv vs Con↑ [18], AD vs Con↓ [19]
8,12-iso-iPF-VI
 AD vs MCI↑ [73]↔[74]
LysoPC (18:1)
ADconv vs Con↑ [18]
 
 MCI vs Con↑ [73]↔[74]
LysoPC (20:0)
ADconv vs Con↑ [18]
Ent-7(RS)-7-F2t-dihomo-IsoP
AD vs Con↑ [75]
LysoPC (20:3)
AD vs Con↓ [19]
17-epi-17-F2t-dihomo-IsoP
AD vs Con↑ [75]
LysoPC (22:0)
ADconv vs Con↑ [18]
15(R)-15-F2t-IsoP
AD vs Con↑ [75]
LysoPC (24:0)
ADconv vs Con↑ [18]
PGF
AD vs Con↓ [75]
PC aa C38:4
HR MCI-AD↑ [50], HR Con-AD↓ [50]
4(RS)-F4t-NeuroP
AD vs Con↑ [75]
PC aa C38:4 (16:0, 22:4)
OR AD↑ [21]
  
PC aa C38:4 (18:0, 20:4)
OR AD↑ [21]
Clusterin glycosylation
 
PC ae C34:2
HR Con-AD↑ [50]
β64N_SA1-(HexNAc-Hex)2-core
 AD low atrophy vs AD high atrophy↑ [76]
PC aa C38:5 (18:0, 20:5)
OR AD↓ [21]
β64N_SA2-(HexNAc-Hex)2-core
AD low atrophy vs AD high atrophy↑ [76
PC aa C40:4 (18:0, 22:4)
OR AD↑ [21]
β64N_SA1-(HexNAc-Hex)3-core
AD low atrophy vs AD high atrophy↑ [76]
PC aa C40:5 (18:0, 22:5)
OR AD↑ [21]
β64N_SA2-(HexNAc-Hex)3-core
AD low atrophy vs AD high atrophy↑ [76]
PC aa C40:5 (18:1, 22:4)
OR AD↑ [21]
β64N_SA1-(HexNAc-Hex)4-core
AD low atrophy vs AD high atrophy↑ [76]
PlsCho (18:0-18:1)
ADconv vs Con↑ [18]
β64N_SA3-(HexNAc-Hex)3-core
AD low atrophy vs AD high atrophy↑ [76]
PlsCho (18:0-22:6)
ADconv vs Con↑ [18]
β64N_SA2-(HexNAc-Hex)4-core
AD low atrophy vs AD high atrophy↑ [76]
Cer16:0
MCI vs AD↓ [51]
β64N_SA3-(HexNAc-Hex)4-core
AD low atrophy vs AD high atrophy↑ [76]
Cer18:0
MCI vs AD↓ [51]
  
Cer20:0
MCI vs AD↓ [51]
Other Metabolites
 
Cer22:0
MCI vs AD↓ [51], MCI vs Con↓ [51]
2-Aminoadipic acid
AD vs Con [48
Cer24:0
MCI vs AD↓ [51], MCI vs Con↓ [51]
3-Hydroxyisovaleric acid
AD vs Con [48
Cer26:0
MCI vs AD↓ [51], MCI vs Con↓ [51]
8-iso-PGF2a (15-F2t-IsoP)
AD vs Con [48]
SM (d18:1/20:1)
AD vs Con [48]
Anthranilic acid
HR AD↑ [64]
SM (d18:1/23:0)
AD vs Con [48]
Asymetric dimethyl-Arginine
AD vs Con↑ [36]
SM (C18:1)
HR MCI-AD↑ [50], HR Con-AD↑ [50]
β-Hydroxy butyric acid
AD vs Con↓ [35], MCI vs Con↓ [35]
SM (C16:0)
HR Con-AD↑ [50]
Choline
AD vs Con↑ [36], MCI vs Con↑ [36]
SM (C16:1)
HR Con-AD↑ [50]
Creatine
AD vs MCI↓ [36], AD vs Con↓ [77]
SM (OH) (C14:1)
HR Con-AD↑ [50]
Creatinine
AD vs con↑ [36]
SM (OH) (C22:1)
OR Dem vs Con↓ [17]
Dihydrosphingosine
AD vs Con↓ [19]
  
Hypoxanthine
HR AD↓ [64]
Bile acids
 
Palmitic amide
AD vs Con↑ [19]
Cholic acid
OR AD vs Con↓ [52]
Ornithine
AD vs Con [48]
 
MCInon-conv vs MCIconv ↓ [52]
S-3-Hyxroxyisobutyric acid
AD vs Con [48]
Deoxycholic acid,
OR AD vs Con↑ [52]
S-adenosylhomocysteine
MCI vs Con↑ [63]
Glucodeoxycholic acid
OR AD vs Con↑ [52]
  
Taurodeoxycholic acid
OR AD vs Con↑ [52]
  
Glycolithocholic acid
OR AD vs Con↑ [52]
  
Taurolithocholic acid
OR AD vs Con↑ [52]
  
AD Alzheimer’s disease, ADconv AD converter, DAG diacylglycerols, Dem dementia, DLB dementia with Lewy bodies, HR hazard ratio, MCI mild cognitive impairment, MCIconv MCI converter, MCInon-conv MCI non-converter, MMA methylmalonic acid, OR odds ratio, Ox. stress-med., oxidative stress-mediated, TG triglycerides, ↑ significantly increased, ↓ significantly decreased, ↔ unchanged
Sphingolipids are another class of lipids which are part of the plasma membrane and involved in a myriad of different biological processes such as signal transduction and cell–cell interaction [23]. Changes of different sphingolipids in AD blood using MS techniques have been reported in single studies and are listed in Table 2.

Apolipoprotein E (ApoE)

ApoE is the most prevalent lipoprotein in the brain and it exists as three major isoforms, ApoE2, ApoE3 and ApoE4 [24]. ApoE4 is the most important genetic risk factor for AD [1] and might be an interesting biomarker candidate. Han and colleagues used two orthogonal methods to measure total ApoE in serum including MRM and observed lower ApoE levels in AD with both methods [25], Two other studies did not find a difference [26, 27]. Inconsistent results of ApoE levels in AD blood were also reported in studies using immunassays [2831]. In addition, total ApoE levels in blood are different between carriers of different ApoE genotypes [26, 27, 32] which renders ApoE quite unsuitable as a disease biomarker. However, it turned out that MRM seems to be a valuable method for ApoE genotyping. Several groups reported the successful isoform identification of ApoE in blood by quantifying allele-specific peptides with up to 100% concordance to classical genotyping [32, 33].

Acylcarnitines/Fatty acids/Di-and Triglycerides

L-Carnitine and acylcarnitines are essential players in energy metabolism but also other functions e.g. in the brain are reported. This includes protein modulation, neuromodulation, protection from excitotoxicity, antioxidant and anti-apoptotic functions all of which can be linked to neurodegenerative diseases [34]. Consistent changes in AD in two independent studies have been reported for five acylcarnitines. Acetyl-carnitine (C2), dodecanoyl-carnitine (C12), dodecenoyl-carnitine (C12:1) and tetradecenyl-carnitine (C14:1) were reduced in plasma [35] and serum [20] of AD patients compared with controls and MCI patients. C2, C12:1 and C14:1 were already reduced in MCI [20]. Both studies used MRM for acylcarnitine measurement. In contrast, octanoyl-carnitine (C8) was increased in AD in two studies analyzing plasma [35] or serum [36] samples. Reports on serum C8 levels in MCI were inconsistent [20, 36]. Changes of other acylcarnitines in blood of AD patients were reported only by single studies and are listed in Table 2. Free fatty acids and diacyl- and triglycerides measured with MS-based methods are also depicted in Table 2 because changes in AD were described in single studies only.
A link between cholesterol and AD comes from epidemiological studies indicating that high cholesterol levels in blood are associated with a higher risk for the development of AD. In addition, cholesterol can influence APP processing and Aβ aggregation [37]. In the brain, cholesterol is converted to the more hydrophilic 24S-hydroxycholesterol to eliminate it from the brain because cholesterol itself cannot cross the blood–brain-barrier. 24S-hydroxycholesterol is an important metabolite in the regulation of cholesterol homeostasis in the brain [38]. Several studies investigated 24S-hydroxycholesterol levels in blood of AD patients using MS but with conflicting results. Lütjohann et al. [39] and Zuliani et al. [40] observed significantly increased 24S-hydroxycholesterol levels in plasma of AD patients compared with controls although with a substantial overlap between groups. In contrast, Solomon and colleagues found reduced levels in AD [41] and another study observed no difference between AD and controls [42]. Thus, 24S-hydroxycholesterol might not be an optimal biomarker candidate for AD. Two precursors of cholesterol synthesis, lanosterol and lathosterol, have consistently been reported to be lower in AD patients in two studies [41, 43] but also with a high variation within groups. This might limit their use as a biomarker. Several other cholesterol-related metabolites such as bile acids, steroids and vitamin D were measured in blood of AD patients in single studies or with conflicting results and they are listed in Table 2.

Other Molecules

Several other molecules were investigated in blood of AD patients using MS methods and were suggested as potential biomarker candidates. For a majority of them, significant changes in AD were reported in single studies only or with conflicting results and are therefore summarized in this section and listed in Table 2. This includes the group of metal ions. Here, consistent changes in AD in more than one study were described only for cobalt (down-regulated) and mercury (up-regulated) [44, 45]. Furthermore, markers of oxidative stress, amino acids and their derivatives and other metabolites or proteins have been described to be affected in blood of AD patients. However, their suitability as AD biomarkers is difficult to assess due to the single reports and also the question of specificity for AD.

Conclusion

To date, MS-based blood biomarkers for AD are still sparse. Although many studies used MS to identify AD biomarker candidates in blood, most candidates were reported in single studies only and need further validation. In addition, fold-changes in AD patients were often low with substantial overlap of blood levels with the control group which makes them unsuitable as a biomarker. There is also the question of specificity, especially for changes of many metabolites such as lipids since short- and long-term nutritional behavior significantly influences blood levels. Fasting before sample collection alone might not account for these effects. Thus, they might be risk factors for AD but not useful biomarker candidates. The most promising MS-based blood biomarkers for AD at the moment seem to be the determination of Aβ peptides by IP- MS [12, 13] and the “genotyping” of ApoE by MRM [33].
The major strength of MS-based techniques is their high specificity for the target analyte and against matrix effects. Current efforts to measure the core AD biomarkers, Aβ42, total-tau and phospho-tau, in blood with immunoassays struggle with the high complexity of this matrix and the low abundance of the proteins. This could be the place for MS techniques, eventually in combination with immunological methods, to overcome this problems as has successfully been shown for Aβ peptides by Nakamura and colleagues [12]. The improvement of the diagnostic performance of tau determination in blood and the so far unsuccessful measurement of a synaptic blood marker for AD could be promising applications of MS in the current biomarker development. Definitely, MS will be essential in the definition of reference methods for the measurement of AD biomarkers in blood.

Acknowledgements

Funding

No funding or sponsorship was received for publication of this article.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Disclosures

Patrick Oeckl and Markus Otto have nothing to disclose.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.
Open Access This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License ( http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​ ), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
1.
Zurück zum Zitat Scheltens P, Blennow K, Breteler MMB, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer’s disease. Lancet. 2016;388:505–17.PubMedCrossRef Scheltens P, Blennow K, Breteler MMB, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer’s disease. Lancet. 2016;388:505–17.PubMedCrossRef
2.
Zurück zum Zitat McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011;7:263–9.CrossRef McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011;7:263–9.CrossRef
3.
4.
Zurück zum Zitat 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-β in cerebrospinal fluid. Clin Chem. 2014;60:987–94.PubMedCrossRef 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-β in cerebrospinal fluid. Clin Chem. 2014;60:987–94.PubMedCrossRef
5.
Zurück zum Zitat 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 Aβ38, Aβ40, and Aβ42 in cerebrospinal fluid of Alzheimer’s disease patients and healthy controls. J Alzheimers Dis. 2013;33:1021–32.PubMedCrossRef 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 Aβ38, Aβ40, and Aβ42 in cerebrospinal fluid of Alzheimer’s disease patients and healthy controls. J Alzheimers Dis. 2013;33:1021–32.PubMedCrossRef
6.
Zurück zum Zitat Pannee J, Törnqvist U, Westerlund A, Ingelsson M, Lannfelt L, Brinkmalm G, et al. The amyloid-β degradation pattern in plasma—a possible tool for clinical trials in Alzheimer’s disease. Neurosci Lett. 2014;573:7–12.PubMedCrossRef Pannee J, Törnqvist U, Westerlund A, Ingelsson M, Lannfelt L, Brinkmalm G, et al. The amyloid-β degradation pattern in plasma—a possible tool for clinical trials in Alzheimer’s disease. Neurosci Lett. 2014;573:7–12.PubMedCrossRef
7.
Zurück zum Zitat Kim JS, Ahn H-S, Cho SM, Lee JE, Kim Y, Lee C. Detection and quantification of plasma amyloid-β by selected reaction monitoring mass spectrometry. Anal Chim Acta. 2014;840:1–9.PubMedCrossRef Kim JS, Ahn H-S, Cho SM, Lee JE, Kim Y, Lee C. Detection and quantification of plasma amyloid-β by selected reaction monitoring mass spectrometry. Anal Chim Acta. 2014;840:1–9.PubMedCrossRef
8.
Zurück zum Zitat Kaneko N, Nakamura A, Washimi Y, Kato T, Sakurai T, Arahata Y, et al. Novel plasma biomarker surrogating cerebral amyloid deposition. Proc Jpn Acad Ser B. 2014;90:353–64.CrossRef Kaneko N, Nakamura A, Washimi Y, Kato T, Sakurai T, Arahata Y, et al. Novel plasma biomarker surrogating cerebral amyloid deposition. Proc Jpn Acad Ser B. 2014;90:353–64.CrossRef
9.
Zurück zum Zitat Kaneko N, Yamamoto R, Sato T-A, Tanaka K. Identification and quantification of amyloid beta-related peptides in human plasma using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Proc Jpn Acad Ser B. 2014;90:104–17.CrossRef Kaneko N, Yamamoto R, Sato T-A, Tanaka K. Identification and quantification of amyloid beta-related peptides in human plasma using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Proc Jpn Acad Ser B. 2014;90:104–17.CrossRef
10.
Zurück zum Zitat Price JL, Morris JC. Tangles and plaques in nondemented aging and preclinical; Alzheimer’s disease. Ann Neurol. 1999;45:358–68.PubMedCrossRef Price JL, Morris JC. Tangles and plaques in nondemented aging and preclinical; Alzheimer’s disease. Ann Neurol. 1999;45:358–68.PubMedCrossRef
11.
Zurück zum Zitat Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018;14:535–62.CrossRef Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018;14:535–62.CrossRef
12.
Zurück zum Zitat Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Doré V, et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature. 2018;554:249–54.PubMedCrossRef Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Doré V, et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature. 2018;554:249–54.PubMedCrossRef
14.
Zurück zum Zitat Kosicek M, Hecimovic S. Phospholipids and Alzheimer’s disease: alterations, mechanisms and potential biomarkers. Int J Mol Sci. 2013;14:1310–22.PubMedPubMedCentralCrossRef Kosicek M, Hecimovic S. Phospholipids and Alzheimer’s disease: alterations, mechanisms and potential biomarkers. Int J Mol Sci. 2013;14:1310–22.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Mapstone M, Cheema AK, Fiandaca MS, Zhong X, Mhyre TR, MacArthur LH, et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nat Med. 2014;20:415–8.PubMedPubMedCentralCrossRef Mapstone M, Cheema AK, Fiandaca MS, Zhong X, Mhyre TR, MacArthur LH, et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nat Med. 2014;20:415–8.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Li D, Misialek JR, Boerwinkle E, Gottesman RF, Sharrett AR, Mosley TH, et al. Prospective associations of plasma phospholipids and mild cognitive impairment/dementia among African Americans in the ARIC Neurocognitive Study. Alzheimer’s Dement Diagnosis, Assess Dis Monit. 2017;6:1–10. Li D, Misialek JR, Boerwinkle E, Gottesman RF, Sharrett AR, Mosley TH, et al. Prospective associations of plasma phospholipids and mild cognitive impairment/dementia among African Americans in the ARIC Neurocognitive Study. Alzheimer’s Dement Diagnosis, Assess Dis Monit. 2017;6:1–10.
17.
Zurück zum Zitat Li D, Misialek JR, Boerwinkle E, Gottesman RF, Sharrett AR, Mosley TH, et al. Plasma phospholipids and prevalence of mild cognitive impairment and/or dementia in the ARIC Neurocognitive Study (ARIC-NCS). Alzheimer’s Dement Diagnosis, Assess Dis Monit. 2016;3:73–82. Li D, Misialek JR, Boerwinkle E, Gottesman RF, Sharrett AR, Mosley TH, et al. Plasma phospholipids and prevalence of mild cognitive impairment and/or dementia in the ARIC Neurocognitive Study (ARIC-NCS). Alzheimer’s Dement Diagnosis, Assess Dis Monit. 2016;3:73–82.
18.
Zurück zum Zitat Dorninger F, Moser AB, Kou J, Wiesinger C, Forss-Petter S, Gleiss A, et al. Alterations in the plasma levels of specific choline phospholipids in Alzheimer’s disease mimic accelerated aging. J Alzheimer’s Dis. 2018;62:841–54.CrossRef Dorninger F, Moser AB, Kou J, Wiesinger C, Forss-Petter S, Gleiss A, et al. Alterations in the plasma levels of specific choline phospholipids in Alzheimer’s disease mimic accelerated aging. J Alzheimer’s Dis. 2018;62:841–54.CrossRef
19.
Zurück zum Zitat Cui Y, Liu X, Wang M, Liu L, Sun X, Ma L, et al. Lysophosphatidylcholine and amide as metabolites for detecting alzheimer disease using ultrahigh-performance liquid chromatography-quadrupole time-of-flight mass spectrometry-based metabonomics. J Neuropathol Exp Neurol. 2014;73:954–63.PubMedCrossRef Cui Y, Liu X, Wang M, Liu L, Sun X, Ma L, et al. Lysophosphatidylcholine and amide as metabolites for detecting alzheimer disease using ultrahigh-performance liquid chromatography-quadrupole time-of-flight mass spectrometry-based metabonomics. J Neuropathol Exp Neurol. 2014;73:954–63.PubMedCrossRef
20.
Zurück zum Zitat Cristofano A, Sapere N, La Marca G, Angiolillo A, Vitale M, Corbi G, et al. Serum levels of acyl-carnitines along the continuum from normal to Alzheimer’s dementia. PLoS One. 2016;11:e0155694.PubMedPubMedCentralCrossRef Cristofano A, Sapere N, La Marca G, Angiolillo A, Vitale M, Corbi G, et al. Serum levels of acyl-carnitines along the continuum from normal to Alzheimer’s dementia. PLoS One. 2016;11:e0155694.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Ritchie SA, Jayasinge D, Wang L, Goodenowe DB. Improved specificity of serum phosphatidylcholine detection based on side-chain losses during negative electrospray ionization tandem mass spectrometry. Anal Bioanal Chem. 2016;408:7811–23.PubMedCrossRef Ritchie SA, Jayasinge D, Wang L, Goodenowe DB. Improved specificity of serum phosphatidylcholine detection based on side-chain losses during negative electrospray ionization tandem mass spectrometry. Anal Bioanal Chem. 2016;408:7811–23.PubMedCrossRef
22.
Zurück zum Zitat Casanova R, Varma S, Simpson B, Kim M, An Y, Saldana S, et al. Blood metabolite markers of preclinical Alzheimer’s disease in two longitudinally followed cohorts of older individuals. Alzheimer’s Dement. 2016;12:815–22.CrossRef Casanova R, Varma S, Simpson B, Kim M, An Y, Saldana S, et al. Blood metabolite markers of preclinical Alzheimer’s disease in two longitudinally followed cohorts of older individuals. Alzheimer’s Dement. 2016;12:815–22.CrossRef
23.
Zurück zum Zitat Hannun YA, Obeid LM. Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol. 2018;19:175–91.PubMedCrossRef Hannun YA, Obeid LM. Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol. 2018;19:175–91.PubMedCrossRef
25.
Zurück zum Zitat Han S-H, Kim JS, Lee Y, Choi H, Kim J-W, Na DL, et al. Both targeted mass spectrometry and flow sorting analysis methods detected the decreased serum apolipoprotein E level in Alzheimer’s disease patients. Mol Cell Proteom. 2014;13:407–19.CrossRef Han S-H, Kim JS, Lee Y, Choi H, Kim J-W, Na DL, et al. Both targeted mass spectrometry and flow sorting analysis methods detected the decreased serum apolipoprotein E level in Alzheimer’s disease patients. Mol Cell Proteom. 2014;13:407–19.CrossRef
26.
Zurück zum Zitat Martínez-Morillo E, Hansson O, Atagi Y, Bu G, Minthon L, Diamandis EP, et al. Total apolipoprotein E levels and specific isoform composition in cerebrospinal fluid and plasma from Alzheimer’s disease patients and controls. Acta Neuropathol. 2014;127:633–43.PubMedCrossRef Martínez-Morillo E, Hansson O, Atagi Y, Bu G, Minthon L, Diamandis EP, et al. Total apolipoprotein E levels and specific isoform composition in cerebrospinal fluid and plasma from Alzheimer’s disease patients and controls. Acta Neuropathol. 2014;127:633–43.PubMedCrossRef
27.
Zurück zum Zitat Simon R, Girod M, Fonbonne C, Salvador A, Clément Y, Lantéri P, et al. Total ApoE and ApoE4 isoform assays in an Alzheimer’s disease case-control study by targeted mass spectrometry (n = 669): a pilot assay for methionine-containing proteotypic peptides. Mol Cell Proteom. 2012;11:1389–403.CrossRef Simon R, Girod M, Fonbonne C, Salvador A, Clément Y, Lantéri P, et al. Total ApoE and ApoE4 isoform assays in an Alzheimer’s disease case-control study by targeted mass spectrometry (n = 669): a pilot assay for methionine-containing proteotypic peptides. Mol Cell Proteom. 2012;11:1389–403.CrossRef
28.
Zurück zum Zitat Gupta VB, Laws SM, Villemagne VL, Ames D, Bush AI, Ellis KA, et al. Plasma apolipoprotein E and Alzheimer disease risk: the AIBL study of aging. Neurology. 2011;76:1091–8.PubMedCrossRef Gupta VB, Laws SM, Villemagne VL, Ames D, Bush AI, Ellis KA, et al. Plasma apolipoprotein E and Alzheimer disease risk: the AIBL study of aging. Neurology. 2011;76:1091–8.PubMedCrossRef
29.
Zurück zum Zitat Taddei K, Clarnette R, Gandy S, Martins RN. Increased plasma apolipoprotein E (apoE) levels in Alzheimer’s disease. Neurosci Lett. 1997;223:29–32.PubMedCrossRef Taddei K, Clarnette R, Gandy S, Martins RN. Increased plasma apolipoprotein E (apoE) levels in Alzheimer’s disease. Neurosci Lett. 1997;223:29–32.PubMedCrossRef
30.
Zurück zum Zitat Scacchi R, Gambina G, Ruggeri M, Martini M, Ferrari G, Silvestri M, et al. Plasma levels of apolipoprotein E and genetic markers in elderly patients with Alzheimer’s disease. Neurosci Lett. 1999;259:33–6.PubMedCrossRef Scacchi R, Gambina G, Ruggeri M, Martini M, Ferrari G, Silvestri M, et al. Plasma levels of apolipoprotein E and genetic markers in elderly patients with Alzheimer’s disease. Neurosci Lett. 1999;259:33–6.PubMedCrossRef
31.
Zurück zum Zitat Slooter AJ, de Knijff P, Hofman A, Cruts M, Breteler MM, Van Broeckhoven C, et al. Serum apolipoprotein E level is not increased in Alzheimer’s disease: the Rotterdam study. Neurosci Lett. 1998;248:21–4.PubMedCrossRef Slooter AJ, de Knijff P, Hofman A, Cruts M, Breteler MM, Van Broeckhoven C, et al. Serum apolipoprotein E level is not increased in Alzheimer’s disease: the Rotterdam study. Neurosci Lett. 1998;248:21–4.PubMedCrossRef
32.
Zurück zum Zitat Blanchard V, Ramin-Mangata S, Billon-Crossouard S, Aguesse A, Durand M, Chemello K, et al. Kinetics of plasma apolipoprotein E isoforms by LC-MS/MS: a pilot study. J Lipid Res. 2018;59:892–900.PubMedPubMedCentralCrossRef Blanchard V, Ramin-Mangata S, Billon-Crossouard S, Aguesse A, Durand M, Chemello K, et al. Kinetics of plasma apolipoprotein E isoforms by LC-MS/MS: a pilot study. J Lipid Res. 2018;59:892–900.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Hirtz C, Vialaret J, Nouadje G, Schraen S, Benlian P, Mary S, et al. Development of new quantitative mass spectrometry and semi-automatic isofocusing methods for the determination of Apolipoprotein E typing. Clin Chim Acta. 2016;454:33–8.PubMedCrossRef Hirtz C, Vialaret J, Nouadje G, Schraen S, Benlian P, Mary S, et al. Development of new quantitative mass spectrometry and semi-automatic isofocusing methods for the determination of Apolipoprotein E typing. Clin Chim Acta. 2016;454:33–8.PubMedCrossRef
34.
Zurück zum Zitat Jones LL, McDonald DA, Borum PR. Acylcarnitines: role in brain. Prog Lipid Res. 2010;49:61–75.PubMedCrossRef Jones LL, McDonald DA, Borum PR. Acylcarnitines: role in brain. Prog Lipid Res. 2010;49:61–75.PubMedCrossRef
35.
Zurück zum Zitat Ciavardelli D, Piras F, Consalvo A, Rossi C, Zucchelli M, Di Ilio C, et al. Medium-chain plasma acylcarnitines, ketone levels, cognition, and gray matter volumes in healthy elderly, mildly cognitively impaired, or Alzheimer’s disease subjects. Neurobiol Aging. 2016;43:1–12.PubMedCrossRef Ciavardelli D, Piras F, Consalvo A, Rossi C, Zucchelli M, Di Ilio C, et al. Medium-chain plasma acylcarnitines, ketone levels, cognition, and gray matter volumes in healthy elderly, mildly cognitively impaired, or Alzheimer’s disease subjects. Neurobiol Aging. 2016;43:1–12.PubMedCrossRef
36.
Zurück zum Zitat González-Domínguez R, García A, García-Barrera T, Barbas C, Gómez-Ariza JL. Metabolomic profiling of serum in the progression of Alzheimer’s disease by capillary electrophoresis-mass spectrometry. Electrophoresis. 2014;35:3321–30.PubMedCrossRef González-Domínguez R, García A, García-Barrera T, Barbas C, Gómez-Ariza JL. Metabolomic profiling of serum in the progression of Alzheimer’s disease by capillary electrophoresis-mass spectrometry. Electrophoresis. 2014;35:3321–30.PubMedCrossRef
38.
Zurück zum Zitat Leoni V. Oxysterols as markers of neurological disease—a review. Scand J Clin Lab Invest. 2009;69:22–5.PubMedCrossRef Leoni V. Oxysterols as markers of neurological disease—a review. Scand J Clin Lab Invest. 2009;69:22–5.PubMedCrossRef
39.
Zurück zum Zitat Lütjohann D, Papassotiropoulos A, Björkhem I, Locatelli S, Bagli M, Oehring RD, et al. Plasma 24S-hydroxycholesterol (cerebrosterol) is increased in Alzheimer and vascular demented patients. J Lipid Res. 2000;41:195–8.PubMedCrossRef Lütjohann D, Papassotiropoulos A, Björkhem I, Locatelli S, Bagli M, Oehring RD, et al. Plasma 24S-hydroxycholesterol (cerebrosterol) is increased in Alzheimer and vascular demented patients. J Lipid Res. 2000;41:195–8.PubMedCrossRef
40.
Zurück zum Zitat Zuliani G, Donnorso MP, Bosi C, Passaro A, Nora ED, Zurlo A, et al. Plasma 24S-hydroxycholesterol levels in elderly subjects with late onset Alzheimer’s disease or vascular dementia: a case-control study. BMC Neurol. 2011;11:121.PubMedPubMedCentralCrossRef Zuliani G, Donnorso MP, Bosi C, Passaro A, Nora ED, Zurlo A, et al. Plasma 24S-hydroxycholesterol levels in elderly subjects with late onset Alzheimer’s disease or vascular dementia: a case-control study. BMC Neurol. 2011;11:121.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Solomon A, Leoni V, Kivipelto M, Besga A, Oksengård AR, Julin P, et al. Plasma levels of 24S-hydroxycholesterol reflect brain volumes in patients without objective cognitive impairment but not in those with Alzheimer’s disease. Neurosci Lett. 2009;462:89–93.PubMedCrossRef Solomon A, Leoni V, Kivipelto M, Besga A, Oksengård AR, Julin P, et al. Plasma levels of 24S-hydroxycholesterol reflect brain volumes in patients without objective cognitive impairment but not in those with Alzheimer’s disease. Neurosci Lett. 2009;462:89–93.PubMedCrossRef
42.
Zurück zum Zitat Iuliano L, Monticolo R, Straface G, Spoletini I, Gianni W, Caltagirone C, et al. Vitamin E and enzymatic/oxidative stress-driven oxysterols in amnestic mild cognitive impairment subtypes and Alzheimer’s disease. J Alzheimers Dis. 2010;21:1383–92.PubMedCrossRef Iuliano L, Monticolo R, Straface G, Spoletini I, Gianni W, Caltagirone C, et al. Vitamin E and enzymatic/oxidative stress-driven oxysterols in amnestic mild cognitive impairment subtypes and Alzheimer’s disease. J Alzheimers Dis. 2010;21:1383–92.PubMedCrossRef
43.
Zurück zum Zitat Kölsch H, Heun R, Jessen F, Popp J, Hentschel F, Maier W, et al. Alterations of cholesterol precursor levels in Alzheimer’s disease. Biochim Biophys Acta. 2010;1801:945–50.PubMedCrossRef Kölsch H, Heun R, Jessen F, Popp J, Hentschel F, Maier W, et al. Alterations of cholesterol precursor levels in Alzheimer’s disease. Biochim Biophys Acta. 2010;1801:945–50.PubMedCrossRef
44.
Zurück zum Zitat Bocca B, Forte G, Petrucci F, Pino A, Marchione F, Bomboi G, et al. Monitoring of chemical elements and oxidative damage in patients affected by Alzheimer’s disease. Ann Ist Super Sanita. 2005;41:197–203.PubMed Bocca B, Forte G, Petrucci F, Pino A, Marchione F, Bomboi G, et al. Monitoring of chemical elements and oxidative damage in patients affected by Alzheimer’s disease. Ann Ist Super Sanita. 2005;41:197–203.PubMed
45.
Zurück zum Zitat Gerhardsson L, Lundh T, Minthon L, Londos E. Metal concentrations in plasma and cerebrospinal fluid in patients with Alzheimer’s disease. Dement Geriatr Cogn Disord. 2008;25:508–15.PubMedCrossRef Gerhardsson L, Lundh T, Minthon L, Londos E. Metal concentrations in plasma and cerebrospinal fluid in patients with Alzheimer’s disease. Dement Geriatr Cogn Disord. 2008;25:508–15.PubMedCrossRef
46.
Zurück zum Zitat Cui Y, Chen X, Liu L, Xie W, Wu Y, Wu Q, et al. Gas chromatography–mass spectrometry analysis of the free fatty acids in serum obtained from patients with Alzheimer’s disease1. Liu F, Lee D-H, Lagoa R, Kumar S, editors. Biomed Mater Eng. 2015;26:S2165–77. Cui Y, Chen X, Liu L, Xie W, Wu Y, Wu Q, et al. Gas chromatography–mass spectrometry analysis of the free fatty acids in serum obtained from patients with Alzheimer’s disease1. Liu F, Lee D-H, Lagoa R, Kumar S, editors. Biomed Mater Eng. 2015;26:S2165–77.
47.
Zurück zum Zitat Whiley L, Sen A, Heaton J, Proitsi P, García-Gómez D, Leung R, et al. Evidence of altered phosphatidylcholine metabolism in Alzheimer’s disease. Neurobiol Aging. 2014;35:271–8.PubMedCrossRef Whiley L, Sen A, Heaton J, Proitsi P, García-Gómez D, Leung R, et al. Evidence of altered phosphatidylcholine metabolism in Alzheimer’s disease. Neurobiol Aging. 2014;35:271–8.PubMedCrossRef
48.
Zurück zum Zitat de Leeuw FA, Peeters CFW, Kester MI, Harms AC, Struys EA, Hankemeier T, et al. Blood-based metabolic signatures in Alzheimer’s disease. Alzheimer’s Dement Diagnosis, Assess Dis Monit. 2017;8:196–207. de Leeuw FA, Peeters CFW, Kester MI, Harms AC, Struys EA, Hankemeier T, et al. Blood-based metabolic signatures in Alzheimer’s disease. Alzheimer’s Dement Diagnosis, Assess Dis Monit. 2017;8:196–207.
49.
Zurück zum Zitat Wood PL, Medicherla S, Sheikh N, Terry B, Phillipps A, Kaye JA, et al. Targeted lipidomics of fontal cortex and plasma diacylglycerols (DAG) in mild cognitive impairment and Alzheimer’s disease: validation of DAG accumulation early in the pathophysiology of Alzheimer’s disease. J Alzheimer’s Dis. 2015;48:537–46.CrossRef Wood PL, Medicherla S, Sheikh N, Terry B, Phillipps A, Kaye JA, et al. Targeted lipidomics of fontal cortex and plasma diacylglycerols (DAG) in mild cognitive impairment and Alzheimer’s disease: validation of DAG accumulation early in the pathophysiology of Alzheimer’s disease. J Alzheimer’s Dis. 2015;48:537–46.CrossRef
50.
Zurück zum Zitat Varma VR, Oommen AM, Varma S, Casanova R, An Y, Andrews RM, et al. Brain and blood metabolite signatures of pathology and progression in Alzheimer disease: a targeted metabolomics study. PLOS Med. 2018;15:e1002482.PubMedPubMedCentralCrossRef Varma VR, Oommen AM, Varma S, Casanova R, An Y, Andrews RM, et al. Brain and blood metabolite signatures of pathology and progression in Alzheimer disease: a targeted metabolomics study. PLOS Med. 2018;15:e1002482.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Mielke MM, Haughey NJ, Bandaru VVR, Schech S, Carrick R, Carlson MC, et al. Plasma ceramides are altered in mild cognitive impairment and predict cognitive decline and hippocampal volume loss. Alzheimers Dement. 2010;6:378–85.PubMedCrossRef Mielke MM, Haughey NJ, Bandaru VVR, Schech S, Carrick R, Carlson MC, et al. Plasma ceramides are altered in mild cognitive impairment and predict cognitive decline and hippocampal volume loss. Alzheimers Dement. 2010;6:378–85.PubMedCrossRef
52.
Zurück zum Zitat MahmoudianDehkordi S, Arnold M, Nho K, Ahmad S, Jia W, Xie G, et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease—an emerging role for gut microbiome. Alzheimer’s Dement. 2019;15:76–92.CrossRef MahmoudianDehkordi S, Arnold M, Nho K, Ahmad S, Jia W, Xie G, et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease—an emerging role for gut microbiome. Alzheimer’s Dement. 2019;15:76–92.CrossRef
53.
Zurück zum Zitat Benussi L, Ghidoni R, Dal Piaz F, Binetti G, Di Iorio G, Abrescia P. The level of 24-hydroxycholesteryl esters is an early marker of Alzheimer’s disease. J Alzheimer’s Dis. 2017;56:825–33.CrossRef Benussi L, Ghidoni R, Dal Piaz F, Binetti G, Di Iorio G, Abrescia P. The level of 24-hydroxycholesteryl esters is an early marker of Alzheimer’s disease. J Alzheimer’s Dis. 2017;56:825–33.CrossRef
54.
Zurück zum Zitat Sato Y, Suzuki I, Nakamura T, Bernier F, Aoshima K, Oda Y. Identification of a new plasma biomarker of Alzheimer’s disease using metabolomics technology. J Lipid Res. 2012;53:567–76.PubMedPubMedCentralCrossRef Sato Y, Suzuki I, Nakamura T, Bernier F, Aoshima K, Oda Y. Identification of a new plasma biomarker of Alzheimer’s disease using metabolomics technology. J Lipid Res. 2012;53:567–76.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Vaňková M, Hill M, Velíková M, Včelák J, Vacínová G, Lukášová P, et al. Reduced sulfotransferase SULT2A1 activity in patients with Alzheimer’s disease. Physiol Res. 2015;64(Suppl 2):S265–73.PubMedCrossRef Vaňková M, Hill M, Velíková M, Včelák J, Vacínová G, Lukášová P, et al. Reduced sulfotransferase SULT2A1 activity in patients with Alzheimer’s disease. Physiol Res. 2015;64(Suppl 2):S265–73.PubMedCrossRef
56.
Zurück zum Zitat Attal-Khemis S, Dalmeyda V, Michot J-L, Roudier M, Morfin R. Increased total 7-hydroxy-dehydroepiandrosterone in serum of patients with Alzheimer’s disease. J Gerontol Ser A Biol Sci Med Sci. 1998;53A:B125–32.CrossRef Attal-Khemis S, Dalmeyda V, Michot J-L, Roudier M, Morfin R. Increased total 7-hydroxy-dehydroepiandrosterone in serum of patients with Alzheimer’s disease. J Gerontol Ser A Biol Sci Med Sci. 1998;53A:B125–32.CrossRef
57.
Zurück zum Zitat Peña-Bautista C, Baquero M, Ferrer I, Hervás D, Vento M, García-Blanco A, et al. Neuropsychological assessment and cortisol levels in biofluids from early Alzheimer’s disease patients. Exp Gerontol. 2019;123:10–6.PubMedCrossRef Peña-Bautista C, Baquero M, Ferrer I, Hervás D, Vento M, García-Blanco A, et al. Neuropsychological assessment and cortisol levels in biofluids from early Alzheimer’s disease patients. Exp Gerontol. 2019;123:10–6.PubMedCrossRef
58.
Zurück zum Zitat Cho S-H, Jung BH, Lee W-Y, Chung BC. Rapid column-switching liquid chromatography/mass spectrometric assay for DHEA-sulfate in the plasma of patients with Alzheimer’s disease. Biomed Chromatogr. 2006;20:1093–7.PubMedCrossRef Cho S-H, Jung BH, Lee W-Y, Chung BC. Rapid column-switching liquid chromatography/mass spectrometric assay for DHEA-sulfate in the plasma of patients with Alzheimer’s disease. Biomed Chromatogr. 2006;20:1093–7.PubMedCrossRef
59.
Zurück zum Zitat Rammouz G, Lecanu L, Aisen P, Papadopoulos V. A lead study on oxidative stress-mediated dehydroepiandrosterone formation in serum: the biochemical basis for a diagnosis of Alzheimer’s disease. J Alzheimer’s Dis. 2011;24:5–16.CrossRef Rammouz G, Lecanu L, Aisen P, Papadopoulos V. A lead study on oxidative stress-mediated dehydroepiandrosterone formation in serum: the biochemical basis for a diagnosis of Alzheimer’s disease. J Alzheimer’s Dis. 2011;24:5–16.CrossRef
60.
Zurück zum Zitat Littlejohns TJ, Henley WE, Lang IA, Annweiler C, Beauchet O, Chaves PHM, et al. Vitamin D and the risk of dementia and Alzheimer disease. Neurology. 2014;83:920–8.PubMedPubMedCentralCrossRef Littlejohns TJ, Henley WE, Lang IA, Annweiler C, Beauchet O, Chaves PHM, et al. Vitamin D and the risk of dementia and Alzheimer disease. Neurology. 2014;83:920–8.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Olsson E, Byberg L, Karlström B, Cederholm T, Melhus H, Sjögren P, et al. Vitamin D is not associated with incident dementia or cognitive impairment: an 18-y follow-up study in community-living old men. Am J Clin Nutr. 2017;105:936–43.PubMedCrossRef Olsson E, Byberg L, Karlström B, Cederholm T, Melhus H, Sjögren P, et al. Vitamin D is not associated with incident dementia or cognitive impairment: an 18-y follow-up study in community-living old men. Am J Clin Nutr. 2017;105:936–43.PubMedCrossRef
62.
Zurück zum Zitat Fonteh AN, Harrington RJ, Tsai A, Liao P, Harrington MG. Free amino acid and dipeptide changes in the body fluids from Alzheimer’s disease subjects. Amino Acids. 2007;32:213–24.PubMedCrossRef Fonteh AN, Harrington RJ, Tsai A, Liao P, Harrington MG. Free amino acid and dipeptide changes in the body fluids from Alzheimer’s disease subjects. Amino Acids. 2007;32:213–24.PubMedCrossRef
63.
Zurück zum Zitat Dayon L, Guiraud SP, Corthésy J, Da Silva L, Migliavacca E, Tautvydaitė D, et al. One-carbon metabolism, cognitive impairment and CSF measures of Alzheimer pathology: homocysteine and beyond. Alzheimers Res Ther. 2017;9:43.PubMedPubMedCentralCrossRef Dayon L, Guiraud SP, Corthésy J, Da Silva L, Migliavacca E, Tautvydaitė D, et al. One-carbon metabolism, cognitive impairment and CSF measures of Alzheimer pathology: homocysteine and beyond. Alzheimers Res Ther. 2017;9:43.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Chouraki V, Preis SR, Yang Q, Beiser A, Li S, Larson MG, et al. Association of amine biomarkers with incident dementia and Alzheimer’s disease in the Framingham Study. Alzheimer’s Dement. 2017;13:1327–36.CrossRef Chouraki V, Preis SR, Yang Q, Beiser A, Li S, Larson MG, et al. Association of amine biomarkers with incident dementia and Alzheimer’s disease in the Framingham Study. Alzheimer’s Dement. 2017;13:1327–36.CrossRef
65.
Zurück zum Zitat Yang H, Lyutvinskiy Y, Soininen H, Zubarev RA. Alzheimer’s disease and mild cognitive impairment are associated with elevated levels of isoaspartyl residues in blood plasma proteins. J Alzheimer’s Dis. 2011;27:113–8.CrossRef Yang H, Lyutvinskiy Y, Soininen H, Zubarev RA. Alzheimer’s disease and mild cognitive impairment are associated with elevated levels of isoaspartyl residues in blood plasma proteins. J Alzheimer’s Dis. 2011;27:113–8.CrossRef
66.
Zurück zum Zitat González-Domínguez R, García-Barrera T, Gómez-Ariza JL. Characterization of metal profiles in serum during the progression of Alzheimer’s disease. Metallomics. 2014;6:292–300.PubMedCrossRef González-Domínguez R, García-Barrera T, Gómez-Ariza JL. Characterization of metal profiles in serum during the progression of Alzheimer’s disease. Metallomics. 2014;6:292–300.PubMedCrossRef
67.
Zurück zum Zitat De Sole P, Rossi C, Chiarpotto M, Ciasca G, Bocca B, Alimonti A, et al. Possible relationship between Al/ferritin complex and Alzheimer’s disease. Clin Biochem. 2013;46:89–93.PubMedCrossRef De Sole P, Rossi C, Chiarpotto M, Ciasca G, Bocca B, Alimonti A, et al. Possible relationship between Al/ferritin complex and Alzheimer’s disease. Clin Biochem. 2013;46:89–93.PubMedCrossRef
68.
Zurück zum Zitat Boström F, Hansson O, Gerhardsson L, Lundh T, Minthon L, Stomrud E, et al. CSF Mg and Ca as diagnostic markers for dementia with Lewy bodies. Neurobiol Aging. 2009;30:1265–71.PubMedCrossRef Boström F, Hansson O, Gerhardsson L, Lundh T, Minthon L, Stomrud E, et al. CSF Mg and Ca as diagnostic markers for dementia with Lewy bodies. Neurobiol Aging. 2009;30:1265–71.PubMedCrossRef
69.
Zurück zum Zitat Bomboi G, Marchione F, Sepe-Monti M, De Carolis A, Bianchi V, Medda E, et al. Correlation between metal ions and clinical findings in subjects affected by Alzheimer’s disease. Ann Ist Super Sanita. 2005;41:205–12.PubMed Bomboi G, Marchione F, Sepe-Monti M, De Carolis A, Bianchi V, Medda E, et al. Correlation between metal ions and clinical findings in subjects affected by Alzheimer’s disease. Ann Ist Super Sanita. 2005;41:205–12.PubMed
70.
Zurück zum Zitat Rembach A, Doecke JD, Roberts BR, Watt AD, Faux NG, Volitakis I, et al. Longitudinal analysis of serum copper and ceruloplasmin in Alzheimer’s disease. J Alzheimer’s Dis. 2013;34:171–82.CrossRef Rembach A, Doecke JD, Roberts BR, Watt AD, Faux NG, Volitakis I, et al. Longitudinal analysis of serum copper and ceruloplasmin in Alzheimer’s disease. J Alzheimer’s Dis. 2013;34:171–82.CrossRef
71.
Zurück zum Zitat Hare DJ, Doecke JD, Faux NG, Rembach A, Volitakis I, Fowler CJ, et al. Decreased plasma iron in Alzheimer’s disease is due to transferrin desaturation. ACS Chem Neurosci. 2015;6:398–402.PubMedCrossRef Hare DJ, Doecke JD, Faux NG, Rembach A, Volitakis I, Fowler CJ, et al. Decreased plasma iron in Alzheimer’s disease is due to transferrin desaturation. ACS Chem Neurosci. 2015;6:398–402.PubMedCrossRef
72.
Zurück zum Zitat Koç ER, Ilhan A, Zübeyde A, Acar B, Gürler M, Altuntaş A, et al. A comparison of hair and serum trace elements in patients with Alzheimer disease and healthy participants. Turkish J Med Sci. 2015;45:1034–9.CrossRef Koç ER, Ilhan A, Zübeyde A, Acar B, Gürler M, Altuntaş A, et al. A comparison of hair and serum trace elements in patients with Alzheimer disease and healthy participants. Turkish J Med Sci. 2015;45:1034–9.CrossRef
73.
Zurück zum Zitat Praticò D, Clark CM, Liun F, Rokach J, Lee VY-M, Trojanowski JQ. Increase of brain oxidative stress in mild cognitive impairment: a possible predictor of Alzheimer disease. Arch Neurol. 2002;59:972–6.PubMedCrossRef Praticò D, Clark CM, Liun F, Rokach J, Lee VY-M, Trojanowski JQ. Increase of brain oxidative stress in mild cognitive impairment: a possible predictor of Alzheimer disease. Arch Neurol. 2002;59:972–6.PubMedCrossRef
74.
Zurück zum Zitat Irizarry MC, Yao Y, Hyman BT, Growdon JH, Praticò D. Plasma F2A isoprostane levels in Alzheimer’s and Parkinson’s disease. Neurodegener Dis. 2007;4:403–5.PubMedCrossRef Irizarry MC, Yao Y, Hyman BT, Growdon JH, Praticò D. Plasma F2A isoprostane levels in Alzheimer’s and Parkinson’s disease. Neurodegener Dis. 2007;4:403–5.PubMedCrossRef
75.
Zurück zum Zitat Peña-Bautista C, Vigor C, Galano J-M, Oger C, Durand T, Ferrer I, et al. Plasma lipid peroxidation biomarkers for early and non-invasive Alzheimer Disease detection. Free Radic Biol Med. 2018;124:388–94.PubMedCrossRef Peña-Bautista C, Vigor C, Galano J-M, Oger C, Durand T, Ferrer I, et al. Plasma lipid peroxidation biomarkers for early and non-invasive Alzheimer Disease detection. Free Radic Biol Med. 2018;124:388–94.PubMedCrossRef
76.
Zurück zum Zitat Liang H-C, Russell C, Mitra V, Chung R, Hye A, Bazenet C, et al. Glycosylation of human plasma clusterin yields a novel candidate biomarker of alzheimer’s disease. J Proteome Res. 2015;14:5063–76.PubMedCrossRef Liang H-C, Russell C, Mitra V, Chung R, Hye A, Bazenet C, et al. Glycosylation of human plasma clusterin yields a novel candidate biomarker of alzheimer’s disease. J Proteome Res. 2015;14:5063–76.PubMedCrossRef
77.
Zurück zum Zitat González-Domínguez R, García-Barrera T, Gómez-Ariza JL. Application of a novel metabolomic approach based on atmospheric pressure photoionization mass spectrometry using flow injection analysis for the study of Alzheimer’s disease. Talanta. 2015;131:480–9.PubMedCrossRef González-Domínguez R, García-Barrera T, Gómez-Ariza JL. Application of a novel metabolomic approach based on atmospheric pressure photoionization mass spectrometry using flow injection analysis for the study of Alzheimer’s disease. Talanta. 2015;131:480–9.PubMedCrossRef
Metadaten
Titel
A Review on MS-Based Blood Biomarkers for Alzheimer’s Disease
verfasst von
Patrick Oeckl
Markus Otto
Publikationsdatum
01.12.2019
Verlag
Springer Healthcare
Erschienen in
Neurology and Therapy / Ausgabe Sonderheft 2/2019
Print ISSN: 2193-8253
Elektronische ISSN: 2193-6536
DOI
https://doi.org/10.1007/s40120-019-00165-4

Weitere Artikel der Sonderheft 2/2019

Neurology and Therapy 2/2019 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.