Open Access

Potential fluid biomarkers for pathological brain changes in Alzheimer's disease: Implication for the screening of cognitive frailty

  • Authors:
    • Qingwei Ruan
    • Grazia D'Onofrio
    • Daniele Sancarlo
    • Antonio Greco
    • Zhuowei Yu
  • View Affiliations

  • Published online on: August 9, 2016     https://doi.org/10.3892/mmr.2016.5618
  • Pages: 3184-3198
  • Copyright: © Ruan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cognitive frailty (CF) overlaps with early neuropathological alterations associated with aging‑related major neurocognitive disorders, including Alzheimer's disease (AD). Fluid biomarkers for these pathological brain alterations allow for early diagnosis in the preclinical stages of AD, and for objective prognostic assessments in clinical intervention trials. These biomarkers may also be helpful in the screening of CF. The present study reviewed the literature and identified systematic reviews of cohort studies and other authoritative reports. The selection criteria for potentially suitable fluid biomarkers included: i) Frequent use in studies of fluid‑derived markers and ii) evidence of novel measurement techniques for fluid‑derived markers. The present study focused on studies that assessed these biomarkers in AD, mild cognitive impairment and non‑AD demented subjects. At present, widely used fluid biomarkers include cerebrospinal fluid (CSF), total tau, phosphorylated tau and amyloid‑β levels. With the development of novel measurement techniques and improvements in understanding regarding the mechanisms underlying aging‑related major neurocognitive disorders, numerous novel biomarkers associated with various aspects of AD neuropathology are being explored. These include specific measurements of Aβ oligomer or monomer forms, tau proteins in the peripheral plasma and CSF, and novel markers of synaptic dysfunction, neuronal damage and apoptosis, neuronal activity alteration, neuroinflammation, blood brain barrier dysfunction, oxidative stress, metabolites, mitochondrial function and aberrant lipid metabolism. The proposed panels of fluid biomarkers may be useful in the early diagnosis of AD, prediction of the progression of AD from preclinical stages to the dementia stage, and the differentiation of AD from non‑AD dementia. In combination with physical frailty, the present study surmised that these biomarkers may also be used as biomarkers for CF, thus contribute to discovering causes and informing interventions for cognitive impairment in individuals with CF.

Introduction

Cognitive frailty (CF) refers to a heterogeneous clinical syndrome found in elderly individuals that excludes those with AD and other types of dementia, and is characterized by concurrent physical frailty and potentially reversible cognitive impairment (1). CF includes reversible and potentially reversible subtypes (2), and may represent a precursor to neurodegenerative processes. Although studies regarding CF biomarkers are scarce, the neuropathological processes overlap with those in individuals with AD and/or other neurodegenerative diseases, and the final outcomes of CF are AD or non-AD dementia. Therefore, excluding biomarkers of physical frailty, other biomarkers, such as amyloid-β (Aβ) accumulation, neurodegeneration or neuronal injury, may be considered biomarkers for CF. Reversible CF occurs in the later stages of preclinical AD or at the pre-mild cognitive impairment (MCI) stage due to other causes, and can be diagnosed based on subjective cognitive decline (SCD) and/or positive biomarkers. Cognitive impairment in potentially reversible CF is comparable to MCI.

Diagnosis of preclinical sporadic AD or other suspected non-Alzheimer pathologies in reversible CF depends on the evidence of pathophysiological alterations in the brain, as demonstrated by established fluid and imaging biomarkers. Neuronal injury or neurodegeneration-associated biomarkers offer chances to predict cognitive impairment progression and prognosis, and to evaluate outcomes of disease-modifying interventions in clinical trials (3). However, there are several challenges, including invasive or expensive detection techniques, and time-consuming detection procedures for the measurement of current biomarkers for disease-induced early neuropathological alterations. The most important challenges associated with current biomarkers are the measurement techniques; for example, current standard enzyme-linked immunosorbent assay (ELISA) methods are insensitive to very low concentrations of biomarkers in cerebrospinal fluid (CSF) and blood (4). Therefore, blood- and urine-based biomarkers may be more attractive biomarkers for the screening of neuropathological alterations in AD and CF. The present study systematically reviewed the advance of fluid biomarkers for the pathological process of AD, which will likely aid in the discovery of biomarkers for CF.

Materials and methods

Literature search

The search strategy was updated according to best practice recommendations (5,6). Literature searches were performed using MEDLINE (between 1996 and April 2015; https://www.nlm.nih.gov/bsd/pmresources.html) and PubMed (between 1990 and November 2015; www.ncbi.nlm.nih.gov/pubmed) databases. The search queries included: i) Blood, ii) cerebrospinal fluid, iii) urine, iv) Alzheimer's disease and v) mild cognitive impairment.

Study selection

A reviewer scrutinized abstracts found by electronic search in order to identify articles meriting a full review. Entire articles were reviewed before data were extracted from pertinent papers.

The inclusion criteria used for the review protocol were as follows: i) Age ≥60 years, ii) diagnosis of AD according to the criteria of the National Institute on Aging-Alzheimer's Association (NIAAA) (7) or diagnosis of MCI according to NIAAA criteria (8) with a confirmed progression to AD assessed by clinical follow-up, and iii) suitable clinical assessments of cognitive and functional impairment, quality of life and clinical evaluations. The exclusion criteria were: i) No English editing (as we lacked resources for translation), ii) diagnosis of non-AD dementia, and iii) MCI that did not progress to AD. No limits were defined on the grounds of disease duration or drug treatment.

Data extraction

A total of 2,243 papers, and 92 additional articles from recent reviews were found. After reviewing the abstracts, 648 papers were obtained on the basis of the aforementioned inclusion/exclusion criteria and duplicates. After screening, 259 papers were reserved, of which 36 reviews and 158 full-text articles were excluded after more in-depth examination, on the basis of the same inclusion/exclusion criteria. Therefore, 65 published studies were considered eligible for the current review (Fig. 1). Data extraction was divided into four categories among the authors by expertise: Potential blood-derived markers of AD and MCI, potential CSF-derived markers of AD and MCI, potential urinary-derived markers of AD and MCI, and cell-based techniques for analyzing tau pathogenicity. Co-authors provided a detailed abstract of all studies describing their strengths and weaknesses, as well as a general evaluation of the category (9,10). The quality of studies was assessed using the Standards for the Reporting of Diagnostic Accuracy Studies in dementia (11).

Results and Discussion

Potential blood-derived markers of AD and MCI

The potential blood-derived biomarkers for AD and MCI are presented in Table I. The ratio of amyloid precursor protein (APP) forms contained in the platelets was significantly decreased in individuals with mild AD, very mild AD and MCI (12), individuals with MCI that progressed to AD dementia (13), and patients with AD that exhibited cognitive decline (14). Compared with patients with non-amnestic MCI, patients with amnestic MCI exhibited increased levels of coated-platelets, which are produced by collagen and thrombin activation. In addition, the increases in coated-platelet levels were associated with an increased risk for the progression to AD (15). There were significantly lower levels of Aβ1-42 and Aβ1-42/1-40 in the plasma of patients with AD compared with patients with non-AD dementia, or in patients with AD-type MCI compared with patients with other types of MCI (16). A cross-sectional study demonstrated that plasma Aβ1-40 levels were independently associated with microvascular brain injury (17). However, plasma Aβ peptide levels only weakly predicted the conversion to AD in normal individuals (18). In addition, plasma Aβ monomer biomarkers were not sensitive enough to discriminate patients with AD-type MCI from patients with early AD, or to distinguish patients with other types of MCI from patients with other types of early dementia.

Table I

Possible blood-derived markers for identifying the risk of MCI and AD.

Table I

Possible blood-derived markers for identifying the risk of MCI and AD.

StudyMethodsIdentified biomarkersAD (n)MCI (n)Control (n)Refs. (n)
Borroni et al, 2003Western blot analysisDecreased baseline platelet APP predicted progression from MCI to AD3013
Zetterberg et al, 2013Tau5, HT7 and BT2 monoclonal antibodiesHigher tau levels in AD54752520
Zhang et al, 2014Post-mortem autopsyDeletion of the AEP gene substantially reduced tau hyperphosphorylation8821
Mapstone et al, 2014LASSO analysisApoE ε4 was similar in MCI and AD18355322
Whiley et al, 2014LC-MS and MLVThree PCs were significantly lower in AD75828423
Marksteiner et al, 2014Multiplex array or ELISA27 vascular-related proteins; NT-proBNP was significantly increased in AD and MCI43274025
Hye et al, 2014Multiplex (xMAP) assays10 plasma proteins (ApoC3, TTR, A1AT, PEDF, CC4, ICAM 1, RANTES, A1AcidG, cystatin C, clusterin) were associated with disease severity and progression47622045226
Lane et al, 2008ELISA, MRIMCI with ApoE ε4 and BCHE-K progressed to AD46427
Soares et al, 2012Multiplex immunoassay panelIncreases in eotaxin 3, pancreatic polypeptide, NT-proBNP and TN-C; decreases in IgM and ApoE levels in patients with AD and MCI; Apo ε3/ε4 or ε4/ε4 carriers had low C-reactive protein and ApoE levels, and high cortisol, interleukin-13, apolipoprotein B and gamma interferon levels973455428
Sattlecker et al, 2014SOMAscanPSA-ACT, pancreatic prohormone, clusterin and fetuin B were related to AD33114921129
Craig-Schapiro et al, 20102-D DIGE LC-MS/MS and ELISAYKL-40, a putative indicator of neuroinflammation, is elevated in AD, and together with Aβ42, has potential prognostic use as a biomarker for preclinical AD48 (with FTD and other dementia)31
Mangialasche et al, 2013HPLCVitamin E measures enchanced the accuracy of sMRI differentiating patients with AD and MCI from cognitively healthy subjects81868633
Squitti et al, 2009Spectrophotometry and immunoturbidimetry assayHigher baseline levels of free copper were correlated with severe cognitive decline in AD and more obvious disabilities at 1 year8136
Pavlopoulos et al, 2013Post-mortem human autopsy and mouse studyRbAp48 decline is responsible for age-related memory loss837
Nettiksimmons et al, 2015ELISA and INNO BIA assaysHigher risk index of blood markers (telomere length, cystatin, serum glucose, C-reactive protein, albumin, IL-6, ApoE ε4 and Aβ42/40) exhibited severe cognitive decline73939
Apostolova et al, 2015Luminex, quantitative polymerase chain reactionApoE genotype, plasma levels of IL-6R and clusterin were useful for predicting brain amyloidosis; ApoE genotype and plasma IL-6R were useful for predicting the conversion of MCI to AD21140

[i] MCI, mild cognitive impairment; AD, Alzheimer's disease; APP, amyloid precursor proteins; ELISA, enzyme-linked immunosorbent assay; MRI, magnetic resonance imaging; ApoE, apolipoprotein E; BCHE-K, butyrylcholinesterase K-variant; 2-D DIGE, 2-D Fluorescence Difference Gel Electrophoresis; LC-MS, liquid chromatography-mass spectrometry; YKL-40, chitinase-3-like-1; Aβ, amyloid-β; FTD, frontotemporal dementia; NT-proBNP, N-terminal pro-brain natriuretic peptide; TNC, tenascin C; IgM, immunoglobulin M; IL-6, interleukin 6; IL-6R, interleukin 6 receptor; HPLC, reverse-phase high-performance liquid chromatography; HT7 and BT2, anti-tau monoclonal antibody; LASSO, ligand activity by surface similarity order; MLV, multiplatform lipidomic validation; PC, phosphatidylcholine; TTR, transthyretin; A1AT, alpha-1 antitrypsin; PEDF, pigment epithelium-derived factor; CC4: complement C4; ICAM-1, intercellular adhesion molecule 1, RANTES, regulated on activation normal T cell expressed and secreted; A1AcidG, alpha-1-acid glycoprotein; PSA-ACT, prostate-specific antigen complexed to α1-antichymotrypsin; AEP, asparagine endopeptodase.

Tau proteins in the serum are potential biomarkers for neuronal degeneration or damage. A novel ultrasensitive digital immunoassay technology, which is referred to as a single-molecule ELISA, is able to detect clinically relevant proteins in the serum at concentrations <10−15 M (19). Compared with the classic ELISA, this ultrasensitive technique is 1,000-fold more sensitive. The lower limit of quantification is 0.02 pg/ml, which is 100 times higher than the plasma levels of tau proteins (~5 pg/ml). This ultrasensitive technique has detected dynamic changes in tau protein in the serum after hypoxic brain injury, and a mean 2-fold increase in AD individuals, however with some overlaps with the levels in patients with MCI and the controls (20). However, rare protein isoforms that result due to cleavage of the tau protein and disease-specific tau phosphorylation are not recognized by currently available assays (21).

Aberrant lipid metabolism is associated with the pathophysiological processes in AD. Some plasma phospholipids may therefore be considered biomarkers for AD. Reductions in the levels of these phospholipids may accurately predict the progression of an individual from normal cognition to MCI or AD within 2 years (22,23). A biomarker panel that detected decreases in the levels of 10 phospholipids in blood plasma that reflect cell membrane integrity may be able to predict which cognitively normal elderly adults will develop MCI and AD with >90% accuracy (22). Early studies have reported that phosphatidylcholine molecules in AD brains are decreased and that their main metabolite, glycerophosphocholine, is increased (23,24). However, some phosphatidylcholines produced by the sequential methylation of another phosphatide, such as phosphatidylethanolamine, were increased in AD brains. It has previously been demonstrated that three phosphatidylcholine molecules were significantly diminished in patients with AD compared with patients with MCI or age-matched individuals (23).

A previous study measured the plasma levels of 27 vascular-related proteins using multiplex assays or ELISA in patients with MCI or AD, and in healthy controls. The results indicated that N-terminal pro-brain natriuretic peptide (NT-proBNT) was significantly increased in patients with MCI or AD, and could be considered a potential biomarker for AD diagnosis and prognosis (25). Multiplex assays were used to measure 26 previously discovered AD-associated plasma biomarkers in plasma samples from patients with AD and MCI, and elderly non-demented individuals from three multicenter cohorts. These assays demonstrated that 10 proteins, namely transthyretin (TTR), clusterin, cystatin C, alpha-1-acid glycoprotein, intercellular adhesion molecule 1, complement C4, pigment epithelium-derived factor (PEDF), alpha-1 antitrypsin, normal T cells, and expressed and secreted Apolipoprotein C3, were strongly associated with the severity and progression of AD. The 10 proteins, plus the Apolipoproteina E (ApoE) genotype, had the greatest predictive power for identifying dementia from prodromal disease (26). In addition, butyrylcholinesterase K-variant alleles had synergistic effects with the APOE ε4 genotype on the conversion of MCI to AD (27). Another study measured the levels of 146 plasma proteins at baseline and after 1 year using a multiplex immunoassay panel; the results verified that the levels of eotaxin 3, pancreatic polypeptide and NT-proBNT were increased in subjects with AD and MCI, which was similar to changes detected in the CSF, and also demonstrated that the Apo ε3/ε4 or ε4/ε4 genotype depended on the biochemical profile (28). Another study quantified 1,001 proteins by SOMAscan, and demonstrated that protein expression levels of prostate-specific antigen complexed to α1-antichymotrypsin, pancreatic prohormone, clusterin and fetuin B were the most strongly associated with AD. Pancreatic prohormone was significantly associated with left entorhinal cortex and hippocampus atrophy, whereas fetuin B was only related to left entorhinal atrophy. Clusterin was significantly related to the rate of cognitive decline (29).

Other potential blood-derived markers include mitochondrial function indicator N-acetylaspartate (NAA), neuroinflammatory indicator chitinase-3-like 1 (YKL-40), stress protein heme oxygenase-1 (HO-1), nutritional biomarkers, histone-binding protein RbAp48, plasma ketone body (KB), and a cumulative risk index based on several pathways associated with cognitive decline for blood-derived markers. NAA levels are coupled to neuronal mitochondrial function and are correlated with Aβ42 in patients with AD (30). The mean plasma and CSF levels of YKL-40 were enhanced in individuals with a clinical dementia rating between 0.5 and 1, and YKL-40 is considered a potential fluid biomarker for preclinical AD (31). HO-1 protein levels in plasma and CSF, and mRNA levels of lymphocyte HO-1 in individuals with sporadic AD are decreased compared with in normal controls, and other subjects with chronic neurological and medical disorders (32). A previous study suggested that plasma levels of α-/γ-tocopherols, or plasma levels of γ-tocotrienols in combination with structural magnetic resonance imaging (sMRI) measures, may be used to differentiate patients with AD or MCI from cognitively healthy individuals, and predict the conversion of MCI to AD after 1 year of follow-up (33). High levels of homocysteine, a risk factor for microvascular impairment, have been associated with alterations in electroencephalographic rhythms in mild AD, but not in MCI subjects, including unselective increases in cortical delta, theta and alpha rhythms (34). Compared with control individuals, plasma levels of non-enzymatic and enzymatic antioxidants were similarly decreased in patients with MCI and AD (35). Baseline higher levels of free copper may be used to predict severe cognitive decline in AD, and a faster and more obvious progression of disability at 1 year (36). Patients with higher levels of free copper combined with hyperlipidemia were also prone to severe cognitive impairment. RbAp48 modifies histone acylation and is associated with age-related memory impairment. Post-mortem measures demonstrated that levels of RbAp48, which are associated with hippocampus-dependent memory deficits in elderly individuals, were decreased in the dentate gyrus. Similar memory deficits were also observed in RbAp48-deficient transgenic mice (37). Unlike the amyloid cascade hypothesis, which suggests that amyloid pathways are the main therapeutic target, the interventions based on the mitochondrial cascade hypothesis, which suggests that mitochondrial function decline triggers AD pathophysiological cascade, are associated with the restoration and maintenance of mitochondrial function. The increase in plasma KB levels induced by ketone ester in several animal models of AD may improve mitochondrial metabolism and postpone the appearance of AD-like pathological alterations (38). Therefore, low plasma KB levels may be a potential biomarker for AD pathology in individuals with preclinical sporadic AD or reversible CF. A cummulative risk index, including blood-derived markers ApoE ε4 and Aβ42/40, telomere length, blood glucose, cystatin, C-reactive protein, interleukin (IL)-6 and albumin (39), may provide valuable predictive information regarding future cognitive trajectories independent of age and baseline cognitive status. It is a question worthy of further research whether this same risk index could also predict CF. In addition, the ApoE genotype, IL-6 receptor and clusterin plasma levels, together with Auditory-Verbal Learning Test and Trails B have been reported to be useful for predicting brain amyloidosis and MCI progression to AD with modest accuracy (40). In genetic analyses of non-familial AD, the ApoE ε4 genotype contributes to the heterogeneity of disease processes associated with sporadic AD (4143). In addition, alongside the ApoE receptor, the low density lipoprotein receptor 5 repeated allele has been reported to be associated with the risk of dementia, and the correlation was more evident in individuals with mixed or vascular dementia (VaD) compared with AD (44).

Potential CSF-derived markers of AD and MCI

The CSF-derived biomarkers for AD and MCI are presented in Table II. The monomer form Aβ42, tau and phosphorylated (p)-tau are widely used to screen AD and MCI caused by AD, and to predict the conversion of preclinical AD and MCI to AD (45,46), independent of the ApoE genotype (43). Among the CSF biomarkers, CSF Aβ42 has been reported to have the best diagnostic accuracy for the discrimination of AD from frontotemporal dementia (47). CSF Aβ42 and florbetapir were inversely correlated across cognitively normal, MCI and AD groups with 86% consistency (48). Abnormal CSF Aβ levels together with abnormal imaging and serum markers exhibited a better predictive value for the earliest stage of AD (49). Individuals with abnormal CSF proteins, particularly abnormalities of both tau and Aβ42 appeared to be simultaneously at greatest risk of cognitive decline (50) and progression from MCI into incipient AD (51). In addition, the p-tau/Aβ42 ratio in CSF could longitudinally predict cognitive performance loss in subjects with MCI (52). Injury markers in CSF, including total (t)-tau and p-tau, could also predict conversion time, from MCI with amyloid pathology, to dementia (53). However, MRI was slightly better at predicting future clinically defined disease stages and functional decline compared with Aβ42, tau or p-tau in the CSF (54). A combination of MRI or fluorodeoxyglucose-positron emission tomography and CSF promote the predictive value of the conversion of MCI to AD (55,56). However, a combination of CSF t-tau/Aβ42 together with sMRI or neuropsychological tests was not superior, as compared with single trail making test-B or sMRI measures of the right entorhinal cortex (57). Unlike the evaluated CSF values of t-tau, p-tau and Aβ42, the order of the annual increase in ventricular volume detected by serial sMRI was AD group>amnestic MCI group>no AD group. Furthermore, the annual increase in ventricular volume was consistent with the annual decrease in general cognitive and functional indices. The alterations in structure and cognitive function were influenced by the ApoE ε4 genotype (58).

Table II

CSF-derived markers for identifying the risk of MCI and AD.

Table II

CSF-derived markers for identifying the risk of MCI and AD.

StudiesMethodsIdentified biomarkersAD (n)MCI (n)Control (n)Refs.
Lautner et al, 2014ELISALow CSF Aβ in AD and MCI was independent of ApoE genotype30928725143
Riemenschneider et al, 2002ELISAAβ42 levels were significantly decreased in patients with MCI that progressed to probable AD and those with progressive MCI compared with subjects with stable MCI102845
Ewers et al, 2015Multiplex Xmap, luminex platformCSF Aβ42 exhibited the best diagnostic accuracy to discriminate AD from FTD, but showed significant overlap with other types of NAD dementia1671725547
Landau et al, 2013Multiplex xMAP, luminex and Florbetapir imageCSF and amyloid-PET measurements of Aβ were consistent in 86% subjects; CSF Aβ abnormality was not regularly detected prior to fibrillar Aβ accumulation2224910348
Nettiksimmons et al, 2010TaqMan, multiplex Xmap, luminex platform, sMRICSF proteins, together with MRI and ApoE ε4, allele may represent the earliest stages of AD11249
Okonkwo et al, 2010Multiplex Xmap, luminex platform, PFAQAbnormal CSF levels of t-tau, p-tau and Aβ42 predicted functional decline and the conversion from control and MCI to AD, but not further prognosis in patients with AD10019511450
Mattsson et al, 2009ELISACSF Aβ42, t-tau, and p-tau could identify incipient AD with good accuracy in patients with MCI52975030451
Laudau et al, 2010ELISAP-tau181p/Aβ_1-42 predicted longitudinal cognitive decline8552
Van Rossum et al, 2012ELISAThe injury markers CSF t-tau and p-tau and hippocampal atrophy could predict further cognitive decline11053
Vemuri et al, 2009STANDT-tau, p-tau and Aβ42 provided better predictive values than either source alone with regards to the conversion from aMCI to AD9819210954
Eckerström et al, 2013ELISA, sMRIThe combination of CSF Aβ42, t-tau, and p-tau, neuropsychological tests and sMRI was the best predictor of dementia212655
Shaffer et al, 2013xMAP luminex platformCombination of imaging and CSF biomarkers could improve the prediction of conversion from MCI to AD9756
Vemuri et al, 2010Multiplex Xmap, Luminex platform, sMRIThere was no significant average change in CSF biomarkers except t-tau; ApoE ε4 genotype did not influence the changes in CSF biomarkers711499258
Sutphen et al, 2015ELISA, PET + PiBLongitudinal CSF biomarker patterns consistent with AD were first detectable during early middle age, and were associated with later amyloid positivity and cognitive decline16985

[i] MCI, mild cognitive impairment; AD, Alzheimer's disease; CSF, cerebrospinal fluid; ELISA, enzyme-linked immunosorbent assay; Aβ, amyloid-β; STAND, structural abnormality index; t-, total; p-, phosphorylated; aMCI, amnestic MCI; sMRI, structural magnetic resonance imaging; ApoE, apolipoprotein E; PFAQ, Pfeffer Functional Activities Questionnaire; PET, positron emission tomography; FTD, frontotemporal dementia; NAD, non-Alzheimer's disease; PiB, Pittsburgh compound B.

The potentially associated CSF-derived biomarkers for AD and MCI are presented in Table III. A previous study reported that the albumin ratio between CSF and serum is normal in patients with AD independent of age, and is increased in patients with cerebrovascular diseases (59). However, an elevation in the ratio was associated with AD and VaD in an 85-year-old population, and was associated with the conversion from non-dementia to dementia across 3 years of follow-up (60). A previous study indicated that age-dependent elevation in the ratio could be observed in ApoE ε4 allele carriers without cognitive impairment (61). An elevation in the albumin ratio was also closely associated with severity in patients with AD with medial temporal atrophy (62). Another study demonstrated that an age-dependent elevation in the ratio could only be detected in the hippocampal CA1 and dentate gyrus regions of cognitively normal subjects (63). Compared with cognitively normal subjects, patients with MCI exhibited an increase in the ratio and the levels of soluble platelet-derived growth factor receptor-β. These biomarkers of neurovascular unit damage appeared earlier than other biomarkers, including CSF Aβ42 and tau. Increased CSF β-secretase (BACE 1) activity in individuals with sporadic AD may be involved in the amyloidogenic process and axonal degeneration (64). Elevated BACE 1 activity in subjects with AD and MCI has been shown to be associated with the ApoE ε4 genotype, and decreased levels of Aβ42 were only observed in ApoE ε4 carriers with MCI (65). CSF levels of the soluble amyloid precursor proteins (sAPP) α and β were similar between AD and healthy controls; however, sAPP β levels were significantly higher in patients with MCI compared with healthy controls (66). Significantly higher levels of sAPP in the CSF were superior to significantly lower levels of CSF Aβ42 with regards to early prediction of the progression from MCI to AD, and the differential diagnosis of AD from MCI and frontotemporal dementia (67). Compared with the stable MCI group and the control group, the levels of sAPP α and β were significantly higher in the AD group and the MCI progression to AD group; the ApoE ε4 allele had no effects on the levels of sAPP α and β (68). CSF Aβ42/Aβ40 values were more sensitive than CSF levels of Aβ42 for the identification of incipient AD in MCI individuals (69). Soluble Aβ oligomers are potential biomarkers for AD; however, compared with the changes in CSF Aβ42 or tau, changes in the levels of soluble oligomers were not superior for the discrimination of AD from controls (70,71). In a comparative study of brain expression of soluble Aβ oligomers, Aβx56, and Aβ trimers and dimers, it was demonstrated that levels of Aβ dimers were highest in subjects with probable AD, and the levels of Aβx56 and Aβ trimers were lowest when compared with age-matched unimpaired and young unimpaired subjects (72). Furthermore, only Aβx56 was correlated with pathological tau proteins and postsynaptic proteins. These results suggested that Aβx56 may contribute to the very early stages of AD pathogenesis. Savage et al reported that higher Aβ oligomer levels predicted more severe dementia (73). However, analogous to the detection of various monomer forms, including Aβ40 and Aβ42, developing even more specific assays to measure the precise nature of oligomers via more sensitive amplification platforms will improve the early diagnostic potential of these biomarkers (74). The amide I band reflects the structural destruction of all types of Aβ peptides. Its downshift in CSF and blood has been detected in patients with MCI that progressed to AD, and its downshift frequency was superior to a single Aβ misfold or the level of specific oligomers (75).

Table III

Potential associated CSF-derived markers for identifying risk of MCI and AD.

Table III

Potential associated CSF-derived markers for identifying risk of MCI and AD.

StudiesMethodsIdentified biomarkersAD (n)MCI (n)Control (n)Refs.
Ewers et al, 2012ELISAThe ApoE ε4 genotype was related to increased BACE 1 activity in AD and MCI subjects60513757
Blennow et al, 1990ELISABBB disturbance is not associated with AD and age, but is associated with vascular factors1185059
Skoog et al, 1998ELISASignificantly higher CSF/serum albumin levels were detected in AD, VaD and women that developed dementia132960
Matsumoto et al, 2007ELISABBB disturbance was related to MTLA, but not to Aβ transport across the BBB4262
Zetterberg et al, 2008Multiplex assayHigher BACE 1 activity in AD and in MCI that progressed to AD871133364
Olsson et al, 2003ELISAThe levels of sAPP α and β did not change between AD and healthy controls, but sAPP β significantly increased in patients with MCI compared to healthy controls81194266
Perneczky et al, 2011ELISAThe levels of sAPP β in the MCI-AD group were significantly higher than in the MCI-NAD and FTD groups2158867
Lewczuk et al, 2012Multiplex assayThe levels of sAPP α and β were significantly higher in the AD and MCI-AD groups compared with the MCI-NAD and control groups; ApoE ε4 allele did not affect the levels of sAPP α and β887115568
Hansson et al, 2007Aβ42/Aβ40 ratio was superior to Aβ42 concentration with regards to identifying incipient AD in MCI13769
Hölttä et al, 2013ELISA + N-terminal monoclonal antibody (82E1)Higher concentrations of Aβ oligomers in the CSF of AD individuals19916514870
Herskovits et al, 2013ELISA + luminex platformHigher concentrations of Aβ oligomers in the CSF of AD individuals201971
Savage et al 2014ELISACorrelation between Aβ40 and oligomers in AD635473
Yang et al, 2015ELISAHigher concentrations of Aβ oligomers in CSF of AD10101074
Kvartsberg et al, 2015Mass spectrometry and ELISAHigh CSF Ng levels in AD and MCI100408076
Portelius et al, 2015In-house immunoassayLevels of neurogranin were AD≥MCI-AD>stable MCI>controls9517311077
Kester et al, 2015A sandwich immunoassayBaseline CSF levels of neurogranin were significantly higher in patients with AD and MCI that progressed to AD compared to controls65613778
Sjögren et al, 2000ELISANFL levels of late-onset AD and FTD were significantly increased compared with controls, which may reflect white matter degeneration211879
Sjögren et al, 2001ELISACSF levels of GAP 43 correlated with CSF-tau and sAPP and were significantly increased in AD compared with controls471280
de la Monte et al, 1997Quantitative enzyme-linked sandwich immunoassayClinical and post-mortem CSF levels of AD7c-NTP are related to AD neurodegeneration and the severity of dementia171181
Tarawneh et al, 2011sMRI, Amyloid imaging with PC-BCSF VILIP-1/Aβ42 predicted future cognitive impairment at least as well as tau/Aβ42 and p-tau181/Aβ429821182
Tarawneh et al, 2012ELISACSF VILIP-1 and VILIP-1/Aβ42 predicted rates of global cognitive decline similar to tau and tau/Aβ426083
Tarawneh et al, 2015ELISACSF VILIP-1 levels predict rates of whole-brain and regional atrophy similar to tau and p-tau181236484
Czech et al, 2012GC-MS and LC-MS/MSIncreased cortisol levels seemed to be related to the progression of AD; increased cysteine associated with decreased uridine was the best paired combination to identify mild AD795186
Liguori et al, 2015Chemistry assays + ELISALactate levels were significantly higher in AD compared with in controls and VaD; higher CSF levels of t-tau and p-tau proteins corresponded to lower lactate levels1458087
Vafadar Isfahani et al, 2012Bioinformatic analysis of proteomic profilesLevels of the biomarker panel, including SPARC-like 1 protein, fibrinogen alpha chain precursor, Aβ, ApoE precursor, serum albumin precursor, keratin type I cytoskeletal 9 and tetranectin were AD>MCI>controls33102088
Wildsmith et al, 2014Targeted proteomicsAmyloid precursor protein, neuronal pentraxin receptor, NrCAM and chromogranin A exhibited significant longitudinal changes in AD4551089
Olsson et al, 2013ELISACSF levels of HFABP were significantly elevated in subjects with AD, and MCI that progressed to AD or VaD961706591
Brys et al, 2009Negative ion chemical ionization gas chromatography/mass spectrometryP-tau (231) is the strongest predictor of the decline from MCI to AD; IP levels uniquely exhibit longitudinal progression effects222194
Ayton et al, 2015Myriad Rules Based Medicine platform, Luminex platformBaseline CSF ferritin levels were negatively associated with cognitive performance, and predicted MCI progression to AD671449195

[i] AD, Alzheimer's disease; MCI, mild cognitive impairment; CSF, cerebrospinal fluid; ELISA, enzyme-linked immunosorbent assay; BBB, blood brain barrier; VaD, vascular dementia; AD7c-NTP, AD-associated neuronal thread protein; NFL, neurofilament; FTD, frontotemporal dementia; GAP 43, growth associated protein 43; sAPP, soluble amyloid precursor proteins; MTLA, medial temporal lobe atrophy; Aβ, amyloid-β; BACE 1, β-secretase; p-, phosphorylated; t-, total; IP, isoprostane; sMRI, structural magnetic resonance imaging; PC-B, Pittsburgh compound-B; VILIP-1, visinin-like protein-1; ApoE, apolipoprotein E; GC-MS, gas chromatography mass-spectrometry; LC-MS/MS, light chromatograpgy MS/MS; HFABP, heart fatty acid binding protein; Ng, neurogranin; NrCAM, neuronal cell adhesion molecule.

Synaptic biomarkers in the CSF are potential early biomarkers for AD. Neurogranin, which is mainly located in dendritic spines, is associated with long-term potentiation and memory consolidation. CSF neurogranin is composed of a series of C-terminal peptides. All neurogranin peptides detected by the ELISA method suggest that CSF neurogranin may be used to monitor synaptic degeneration and reflect the rate of cognitive decline in individuals with prodromal AD (76). Even if it is very rare in CSF, the levels of CSF neurogranin peptide 48–76 exhibit the most obvious increase in patients with AD. A progressive decline in CSF levels of neurogranin could be observed from individuals with AD, MCI that progressed to AD, stable MCI to controls (77). Patients with AD or MCI that progressed to AD demonstrated significantly higher baseline CSF levels of neurogranin compared with stable MCI or controls (77,78). CSF levels of the cytoskeleton light neurofilament protein were significantly higher in patients with late-onset AD or frontotemporal dementia compared with controls (79). In addition, alterations in the CSF levels of axonal growth-associated protein-43 were associated with changes in CSF tau and sAPP levels, and patients with AD exhibited a significant increase compared with controls (80). However, the detection of specific peptides and other biomarkers for synaptic dysfunction will benefit from the development of novel ultrasensitive assays.

The onset of cognitive decline is estimated to lag AD pathology by 10–15 years. By the time of the appearance of clinically detectable cognitive impairment, substantial neuronal loss has occurred. As a marker of neuronal damage, the AD-associated neuronal thread protein AD7c-NTP is a ~41 kD membrane-spanning phosphoprotein. The increase in immunoreactive AD7c-NTP in the brain is associated with the increase in p-tau-immunoreactive cytoskeletal lesions, but not the increase in Aβ accumulation. The protein may be involved in neuronal apoptosis and neurite sprouting, and in the pathological alterations of AD. Dying cells in the brain are able to secrete or release the protein into the CSF. Post-mortem CSF levels of AD7c-NTP in AD were significantly higher compared with in age-matched controls. The CSF levels of patients with probable AD were also significantly higher compared with controls or other neurological disease controls, and were associated with the severity of cognitive impairment (81). The CSF levels of visinin-like protein-1 (VILIP-1) and the ratios of VILIP-1/Aβ42, similar to tau and tau/Aβ, may predict future cognitive decline in cognitively normal subjects, differentiate patients with AD from non-AD dementia and healthy controls (82), and predict rates of global cognitive decline in individuals with early AD (83). CSF VILIP-1 levels may also predict rates of whole-brain and regional atrophy with a similar power to CSF levels of tau and p-tau181 in individuals with very mild AD or preclinical AD (84). Longitudinal CSF biomarker patterns, including low Aβ42 in early middle age, markedly increased t-tau, p-tau and VILIP-1 in mid- and late middle age, and increased levels of the neuroinflammatory marker YKL-40 throughout middle age, were useful for screening middle-aged, asymptomatic individuals with AD (85).

Systematic analysis of metabolite profiling of the CSF by magnetic resonance spectroscopy demonstrated that increased cortisol and cysteine levels, and decreased uridine levels, may be involved in the progression of AD. Individuals with severe AD exhibited increased cortisol levels, and individuals with mild AD [Mini Mental State Examination (MMSE)>22] exhibited increased cysteine and decreased uridine levels. Specificity and sensitivity >75% could be obtained for the paired combination of cysteine and uridine to identify mild AD (86). Lactate is a product of glycolytic metabolism; CSF levels of lactate in patients with AD were significantly higher compared with in controls and VaD controls (87). Patients with mild AD exhibited higher lactate levels compared with in moderate and severe AD, and higher lactate levels corresponded with lower levels of t-tau and p-tau. A biomarker panel that analyzed the levels of seven proteins in the CSF by proteomic analysis and mass spectroscopy was able to classify AD individuals from controls with an accuracy of 84.5% (sensitivity 93.3%, specificity 75.7%) (88). Target proteomic analysis detected another four proteins that could reflect obvious longitudinally dynamic alterations during AD progression (89). Other potential CSF-derived markers include α-synuclein, heart fatty acid binding protein (HFABP), PEDF, complexed prostaglandin-d-synthase (PDS) and TTR, isoprostane and ferritin. CSF α-synuclein levels were not only significantly increased in patients with MCI and AD compared with controls, the increase in CSF α-synuclein levels was also significantly associated with the decrease in MMSE scores (90). CSF α-synuclein levels only offered modest sensitivity and specificity as a diagnostic marker of AD. Significantly increased CSF levels of HFABP were detected in patients with AD compared with controls, and in patients with MCI that progressed to AD or VaD when compared with individuals with stable MCI (91). However, HFABP had a lower predictive value than Aβ42, t-tau and p-tau in identifying the progression of MCI to AD and VaD. Cortical neurons and astrocytes in AD brains demonstrated strong immunostaining of PEDF, and CSF PEDF as a biomarker may improve the diagnosis of AD (92). Complexed PDS/TTR exhibited a significant increase in post-mortem ventricular CSF in MCI and late-stage AD compared with diseased control subjects, and lumbar CSF levels of the complex showed a six-fold increase in living subjects with probable AD compared with normal control subjects (93). CSF levels of isoprostane were shown to possess robust longitudinal effects on the progression of MCI to AD. The annual rate of isoprostane was significantly different in the following order: MCI that progressed to AD group>stable MCI group>controls (94). The elevated CSF levels of ferritin were not only associated with cognitive performance but could also predict AD progression (95).

Potential urinary-derived markers for AD and MCI

The potential urinary-derived biomarkers for AD and MCI are presented in Table IV. Patients with AD demonstrated elevated levels of AD7c-NTP in CSF and urine, and the elevated levels of AD7c-NTP were associated with the severity of dementia, i.e., very mild, mild or moderately severe AD. Several studies have suggested that AD7c-NTP is a useful biomarker, and may be widely used to screen the risk of elderly individuals with MCI and AD (9699).

Table IV

Possible urinary-derived markers for identifying risk of MCI and AD.

Table IV

Possible urinary-derived markers for identifying risk of MCI and AD.

StudyMethodsIdentified biomarkersAD (n)MCI (n)Control (n)Refs.
Ghanbari et al, 1998ELISAHigher urinary AD7c-NTP in AD6613496
Ma et al, 2015ELISAHigher AD7c-NTP levels in MCI45606599
Zengi et al, 2011HPLC-ECDUrinary 8-OHdG levels and serum PON1 activity could be used to determine and monitor the status of patients with AD2120100
Kim et al, 2004GC-MSPGF (2 alpha) was increased in AD3420101
Yoshida et al, 2015LC-MS/MS + ELISA3-HPMA/Cre was the most reliable biochemical marker to distinguish AD from MCI322274102
Rabassa et al, 2015F-C assayHigh concentrations of polyphenols were associated with a lower risk of cognitive decline652 (demented-free)103

[i] AD, Alzheimer's disease; MCI, mild cognitive impairment; ELISA, enzyme-linked immunosorbent assay; AD7c-NTP, AD-associated neuronal thread protein; GC-MS, gas chromatography-mass spectrometry; PGF (2 alpha), prostaglandin F (2 alpha); HPLC-ECD, high-performance liquid chromatography-electrochemical detection; 8-OHdG, 8-hydroxy-2′-deoxyguanosine; PON1, paraoxonase/arylesterase 1; LC-MS/MS, liquid chromatography-MS/MS; 3-HPMA/Cre, 3-hydroxypropyl mercapturic acid/creatinine; F-C, Folin-Ciocalteu.

Oxidative stress and oxidative DNA damage have important roles in the process of AD. A major product of oxidative DNA damage is 8-hydroxy-2′-deoxyguanosine (8-OHdG). The antioxidant enzyme paraoxonase 1 (PON1) is able to prevent the oxidation of low-density lipoproteins. Patients with AD exhibited significantly elevated levels of 8-OHdG in urine and significantly decreased PON1 activity in serum, as compared with in healthy elderly volunteers (100). Isoprostanes, which are products of arachidonic acid peroxidation by free radicals, are also biomarkers for oxidative injury. Urinary levels of F2-isoprostanes are significantly elevated in AD subjects (101). In addition, 3-hydroxypropyl mercapturic acid/creatinine reduction in urine is not only associated with stroke but is also an ideal biomarker for differentiating patients with AD from patients with MCI (102). Furthermore, high levels of urinary polyphenols may exert protective effects of polyphenol intake against cognitive impairment, and are associated with a lower risk of obvious cognitive decline in elderly individuals over a 3-year follow-up period (103).

Cell-based techniques for analyzing tau pathogenicity

Recently, two novel methods to detect tau pathogenicity have been developed. One method detects tau seeding activity in various AD brain lysates using a cell-based biosensor assay. In vitro studies have reported that pathogenic tau oligomers can move between cells similar to mechanisms of prion pathogenesis, this is referred to as transcellular spread (104). Briefly, biosensor cells are generated to express human tau proteins fused with cyan fluorescent protein (CFP) or yellow fluorescent protein (YFP); when pathogenic tau from AD samples is added to a culture of biosensor cells, which contain tau proteins fused with CFP or YFP, pathogenic tau promotes the aggregation of two fused proteins and results in a positive fluorescence resonance energy transfer signal (105). The signal intensity reflects the pathogenic tau seeding activity of the AD sample. Detection of pathogenic tau seeding activity from human brain samples could reliably differentiate AD from Huntington's disease and aged controls (106). In addition, P301S tau transgenic mice demonstrated that tau seeding activity is an ideal biomarker of tauopathy. Increases in tau seeding activity occurred 1.5 months earlier than tau deposition. Future verification of pathogenic tau seeding activity in human CSF or blood will improve the diagnostic accuracy of tauopathies in a clinical setting, and provide a noninvasive biomarker to evaluate the therapeutic efficacy of tau-modifying agents in future clinical trials.

The second method quantitatively detects tau pathogenicity in biological samples using monoclonal antibodies against seeding tau. The antibodies can also be used as an immunotherapy that traps pathogenic tau and prevents transcellular spread. Anti-pathogenic tau antibodies that inhibit the ability for tau pathogenicity in vitro could markedly reduce the levels of p-tau, aggregated and insoluble tau, and inhibit microglial activation and improve cognition in P301S tau transgenic mice (107). If these antibodies can be used to quantify tau pathogenicity in CSF and blood from different individuals, they may be considered potentially noninvasive biomarkers for detecting tau pathology.

Implications for CF screening

Aging-related major neurocognitive disorders, particularly AD-induced neurodegeneration, can progress over decades before clinical symptoms become apparent. Biomarkers for brain pathological processes allow for their early diagnosis in preclinical stages and for the development of objective prognostic assessments in clinical intervention trials. Some fluid biomarkers, including Aβ42, t-tau and p-tau, have been widely used in clinical practice and clinical trials. The development of novel measurement techniques greatly promotes the production of novel biomarkers and improves the accuracy of old biomarkers (19,20,73,74). Several novel biomarkers associated with different aspects of AD neuropathology are being developed, including those related to synaptic dysfunction, neuronal damage and apoptosis, neuronal activity alteration, neuroinflammation, oxidative stress, metabolites, mitochondrial function and aberrant lipid metabolism. A bioinformatics approach for identifying specific single nucleotide polymorphisms (108110) and epigenetic markers, including microRNAs (111) in the CSF or blood of patients with MCI, AD or non-AD dementia may also be useful for AD diagnosis and differentiation.

Based on the ordering of AD biomarkers, particularly CSF biomarkers (Aβ42, t-tau and p-tau), and AD-associated biomarkers, such as neural injury and neuroinflammation biomarkers, we classified which one was the earliest event resulting in the heterogeneity of cognitive impairment. These biomarkers may be helpful for the diagnosis of CF and the differential diagnosis of cognitive impairment from CF, including AD or VaD. CF consists of reversible and potentially reversible cognitive impairment subtypes (112). The former is based on SCD, and the latter is comparable to MCI. The most common cause of cognitive impairment of CF is AD. The severity of SCD has been associated with biomarkers for AD in subjects with MCI, including low CSF Aβ42 and high CSF tau or p-tau levels (46). Longitudinal CSF biomarkers, such as Aβ42 reductions in early middle age; markedly increased tau, p-tau and VILIP-1 in mid- and late middle age, and increases in YKL-40 throughout middle age were associated with the severity of cognitive impairment in prelinical AD (85). The combination of fluid biomarkers and imaging biomarkers could further improve diagnostic accuracy of cognitive impairment. In addition, physical frailty and AD may share similar pathophysiological mechanisms. Certain AD-associated fluid biomarkers, such as oxidative stress and inflammatory markers, may also contribute to the screening of physical frailty in CF subjects (112,113). Furthermore, the dynamic changes of AD-specific and AD-associated fluid biomarkers may be helpful for the screening of candidate drugs that affect cognitive impairment and physical frailty.

Acknowledgments

The present study was supported by grants from the Shanghai Hospital Development Center (grant no. SHDC12014221) and the Shanghai Key Laboratory of Clinical Geriatric Medicine (grant no. 13dz2260700).

References

1 

Kelaiditi E, Cesari M, Canevelli M, van Kan GA, Ousset PJ, Gillette-Guyonnet S, Ritz P, Duveau F, Soto ME, Provencher V, et al: Cognitive frailty: Rational and definition from an (I.A.N.A./I.A.G.G.) international consensus group. J Nutr Health Aging. 17:726–734. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Ruan Q, Yu Z, Chen M, Bao Z, Li J and He W: Cognitive frailty, a novel target for the prevention of elderly dependency. Ageing Res Rev. 20:1–10. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Buerger K, Frisoni G, Uspenskaya O, Ewers M, Zetterberg H, Geroldi C, Binetti G, Johannsen P, Rossini PM, Wahlund LO, et al: Validation of Alzheimer's disease CSF and plasma biological markers: The multicentre reliability study of the pilot European Alzheimer's disease neuroimaging initiative (E-ADNI). Exp Gerontol. 44:579–585. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Yang T, Hong S, O'Malley T, Sperling RA, Walsh DM and Selkoe DJ: New ELISAs with high specificity for soluble oligomers of amyloid β-protein detect natural Aβ oligomers in human brain but not CSF. Alzheimers Dement. 9:99–112. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Dixon-Woods M, Agarwal S, Jones D, Young B and Sutton A: Synthesising qualitative and quantitative evidence: A review of possible methods. J Health Serv Res Policy. 10:45–53. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Hannes K and Macaitis K: A move to more systematic and transparent approaches in qualitative evidence synthesis: Update on a review of published papers. Qual Res. 12:402–442. 2012. View Article : Google Scholar

7 

McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr, Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, 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. Alzheimers Dement. 7:263–269. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, et al: The diagnosis of mild cognitive impairment due to Alzheimer's disease: Recommendations from the national institute on Aging-Alzheimer's association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 7:270–279. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Barnett-Page E and Thomas J: Methods for the synthesis of qualitative research: A critical review. BMC Med Res Methodol. 9:592009. View Article : Google Scholar : PubMed/NCBI

10 

Grant MJ and Booth A: A typology of reviews: An analysis of 14 review types and associated methodologies. Health Info Libr J. 26:91–108. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Noel-Storr AH, McCleery JM, Richard E, Ritchie CW, Flicker L, Cullum SJ, Davis D, Quinn TJ, Hyde C, Rutjes AW, et al: Reporting standards for studies of diagnostic test accuracy in dementia: The STARDdem Initiative. Neurology. 83:364–373. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Padovani A, Borroni B, Colciaghi F, Pettenati C, Cottini E, Agosti C, Lenzi GL, Caltagirone C, Trabucchi M, Cattabeni F and Di Luca M: Abnormalities in the pattern of platelet amyloid precursor protein forms in patients with mild cognitive impairment and Alzheimer disease. Arch Neurol. 59:71–75. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Borroni B, Colciaghi F, Caltagirone C, Rozzini L, Broglio L, Cattabeni F, Di Luca M and Padovani A: Platelet amyloid precursor protein abnormalities in mild cognitive impairment predict conversion to dementia of Alzheimer type: A 2-year follow-up study. Arch Neurol. 60:1740–1744. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Baskin F, Rosenberg RN, Iyer L, Hynan L and Cullum CM: Platelet APP isoform ratios correlate with declining cognition in AD. Neurology. 54:1907–1909. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Prodan CI, Ross ED, Stoner JA, Cowan LD, Vincent AS and Dale GL: Coated-platelet levels and progression from mild cognitive impairment to Alzheimer disease. Neurology. 76:247–252. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Lewczuk P, Kornhuber J, Vanmechelen E, Peters O, Heuser I, Maier W, Jessen F, Bürger K, Hampel H, Frölich L, et al: Amyloid beta peptides in plasma in early diagnosis of Alzheimer's disease: A multicenter study with multiplexing. Exp Neurol. 223:366–370. 2010. View Article : Google Scholar

17 

Gurol ME, Irizarry MC, Smith EE, Raju S, Diaz-Arrastia R, Bottiglieri T, Rosand J, Growdon JH and Greenberg SM: Plasma beta-amyloid and white matter lesions in AD, MCI, and cerebral amyloid angiopathy. Neurology. 66:23–29. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Lopez OL, Kuller LH, Mehta PD, Becker JT, Gach HM, Sweet RA, Chang YF, Tracy R and DeKosky ST: Plasma amyloid levels and the risk of AD in normal subjects in the cardiovascular health study. Neurology. 70:1664–1671. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Rissin DM, Kan CW, Campbell TG, Howes SC, Fournier DR, Song L, Piech T, Patel PP, Chang L, Rivnak AJ, et al: Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat Biotechnol. 28:595–599. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J and Hansson O: Plasma tau levels in Alzheimer's disease. Alzheimers Res Ther. 5:92013. View Article : Google Scholar : PubMed/NCBI

21 

Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, Seyfried NT, Hu WT, Liu Z, Wang JZ, et al: Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer's disease. Nat Med. 20:1254–1262. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Mapstone M, Cheema AK, Fiandaca MS, Zhong X, Mhyre TR, MacArthur LH, Hall WJ, Fisher SG, Peterson DR, Haley JM, et al: Plasma phospholipids identify antecedent memory impairment in older adults. Nat Med. 20:415–418. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Whiley L, Sen A, Heaton J, Proitsi P, García-Gómez D, Leung R, Smith N, Thambisetty M, Kloszewska I, Mecocci P, et al: Evidence of altered phosphatidylcholine metabolism in Alzheimer's disease. Neurobiol Aging. 35:271–278. 2014. View Article : Google Scholar

24 

Nitsch RM, Blusztajn JK, Pittas AG, Slack BE, Growdon JH and Wurtman RJ: Evidence for a membrane defect in Alzheimer disease brain. Proc Natl Acad Sci USA. 89:1671–1675. 1992. View Article : Google Scholar : PubMed/NCBI

25 

Marksteiner J, Imarhiagbe D, Defrancesco M, Deisenhammer EA, Kemmler G and Humpel C: Analysis of 27 vascular-related proteins reveals that NT-proBNP is a potential biomarker for Alzheimer's disease and mild cognitive impairment: A pilot-study. Exp Gerontol. 50:114–121. 2014. View Article : Google Scholar

26 

Hye A, Riddoch-Contreras J, Baird AL, Ashton NJ, Bazenet C, Leung R, Westman E, Simmons A, Dobson R, Sattlecker M, et al: Plasma proteins predict conversion to dementia from prodromal disease. Alzheimers Dement. 10:799–807.e2. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Lane R, Feldman HH, Meyer J, He Y, Ferris SH, Nordberg A, Darreh-Shori T, Soininen H, Pirttilä T, Farlow MR, et al: Synergistic effect of apolipoprotein E epsilon4 and butyrylcholinesterase K-variant on progression from mild cognitive impairment to Alzheimer's disease. Pharmacogenet Genomics. 18:289–298. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Soares HD, Potter WZ, Pickering E, Kuhn M, Immermann FW, Shera DM, Ferm M, Dean RA, Simon AJ, Swenson F, et al: Plasma biomarkers associated with the apolipoprotein E genotype and Alzheimer disease. Arch Neurol. 69:1310–1317. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Sattlecker M, Kiddle SJ, Newhouse S, Proitsi P, Nelson S, Williams S, Johnston C, Killick R, Simmons A, Westman E, et al: Alzheimer's disease biomarker discovery using SOMAscan multiplexed protein technology. Alzheimers Dement. 10:724–734. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Jessen F, Lewczuk P, Gür O, Block W, Ende G, Frölich L, Hammen T, Arlt S, Kornhuber J, Kucinski T, et al: Association of N-acetylaspartate and cerebrospinal fluid Aβ42 in dementia. J Alzheimers Dis. 27:393–399. 2011.

31 

Craig-Schapiro R, Perrin RJ, Roe CM, Xiong C, Carter D, Cairns NJ, Mintun MA, Peskind ER, Li G, Galasko DR, et al: YKL-40: A novel prognostic fluid biomarker for preclinical Alzheimer's disease. Biol Psychiatry. 68:903–912. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Schipper HM, Chertkow H, Mehindate K, Frankel D, Melmed C and Bergman H: Evaluation of heme oxygenase-1 as a systemic biological marker of sporadic AD. Neurology. 54:1297–1304. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Mangialasche F, Westman E, Kivipelto M, Muehlboeck JS, Cecchetti R, Baglioni M, Tarducci R, Gobbi G, Floridi P, Soininen H, et al: Classification and prediction of clinical diagnosis of Alzheimer's disease based on MRI and plasma measures of α-/γ-tocotrienols and γ-tocopherol. J Intern Med. 273:602–621. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Babiloni C, Bosco P, Ghidoni R, Del Percio C, Squitti R, Binetti G, Benussi L, Ferri R, Frisoni G, Lanuzza B, et al: Homocysteine and electroencephalographic rhythms in Alzheimer disease: A multicentric study. Neuroscience. 145:942–954. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Rinaldi P, Polidori MC, Metastasio A, Mariani E, Mattioli P, Cherubini A, Catani M, Cecchetti R, Senin U and Mecocci P: Plasma antioxidants are similarly depleted in mild cognitive impairment and in Alzheimer's disease. Neurobiol Aging. 24:915–919. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Squitti R, Bressi F, Pasqualetti P, Bonomini C, Ghidoni R, Binetti G, Cassetta E, Moffa F, Ventriglia M, Vernieri F and Rossini PM: Longitudinal prognostic value of serum 'free' copper in patients with Alzheimer disease. Neurology. 72:50–55. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Pavlopoulos E, Jones S, Kosmidis S, Close M, Kim C, Kovalerchik O, Small SA and Kandel ER: Molecular mechanism for age-related memory loss: The histone-binding protein RbAp48. Sci Transl Med. 5:200ra1152013. View Article : Google Scholar : PubMed/NCBI

38 

Hertz L, Chen Y and Waagepetersen HS: Effects of ketone bodies in Alzheimer's disease in relation to neural hypometabolism, β-amyloid toxicity, and astrocyte function. J Neurochem. 134:7–20. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Nettiksimmons J, Ayonayon H, Harris T, Phillips C, Rosano C, Satterfield S and Yaffe K; Health ABC Study: Development and validation of risk index for cognitive decline using blood-derived markers. Neurology. 84:696–702. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Apostolova LG, Hwang KS, Avila D, Elashoff D, Kohannim O, Teng E, Sokolow S, Jack CR, Jagust WJ, Shaw L, et al: Brain amyloidosis ascertainment from cognitive, imaging, and peripheral blood protein measures. Neurology. 84:729–737. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Smits LL, Pijnenburg YA, van der Vlies AE, Koedam EL, Bouwman FH, Reuling IE, Scheltens P and van der Flier WM: Early onset APOE E4-negative Alzheimer's disease patients show faster cognitive decline on non-memory domains. Eur Neuropsychopharmacol. 25:1010–1017. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Michaelson DM: APOE ε4: The most prevalent yet understudied risk factor for Alzheimer's disease. Alzheimers Dement. 10:861–868. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Lautner R, Palmqvist S, Mattsson N, Andreasson U, Wallin A, Pålsson E, Jakobsson J, Herukka SK, Owenius R, Olsson B, et al: Apolipoprotein E genotype and the diagnostic accuracy of cerebrospinal fluid biomarkers for Alzheimer disease. JAMA Psychiatry. 71:1183–1191. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Helbecque N, Berr C, Cottel D, Fromentin-David I, Sazdovitch V, Ricolfi F, Ducimetière P, Di Menza C and Amouyel P: VLDL receptor polymorphism, cognitive impairment, and dementia. Neurology. 56:1183–1188. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Riemenschneider M, Lautenschlager N, Wagenpfeil S, Diehl J, Drzezga A and Kurz A: Cerebrospinal fluid tau and beta-amyloid 42 proteins identify Alzheimer disease in subjects with mild cognitive impairment. Arch Neurol. 59:1729–1734. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Wolfsgruber S, Jessen F, Koppara A, Kleineidam L, Schmidtke K, Frölich L, Kurz A, Schulz S, Hampel H, Heuser I, et al: Subjective cognitive decline is related to CSF biomarkers of AD in patients with MCI. Neurology. 84:1261–1268. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Ewers M, Mattsson N, Minthon L, Molinuevo JL, Antonell A, Popp J, Jessen F, Herukka SK, Soininen H, Maetzler W, et al: CSF biomarkers for the differential diagnosis of Alzheimer's disease: A large-scale international multicenter study. Alzheimers Dement. 11:1306–1315. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Landau SM, Lu M, Joshi AD, Pontecorvo M, Mintun MA, Trojanowski JQ, Shaw LM and Jagust WJ; Alzheimer's Disease Neuroimaging Initiative: Comparing positron emission tomography imaging and cerebrospinal fluid measurements of β-amyloid. Ann Neurol. 74:826–836. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Nettiksimmons J, Harvey D, Brewer J, Carmichael O, DeCarli C, Jack CR Jr, Petersen R, Shaw LM, Trojanowski JQ, Weiner MW, et al: Subtypes based on cerebrospinal fluid and magnetic resonance imaging markers in normal elderly predict cognitive decline. Neurobiol Aging. 31:1419–1428. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Okonkwo OC, Alosco ML, Griffith HR, Mielke MM, Shaw LM, Trojanowski JQ and Tremont G; Alzheimer's Disease Neuroimaging Initiative: Cerebrospinal fluid abnormalities and rate of decline in everyday function across the dementia spectrum: Normal aging, mild cognitive impairment and Alzheimer disease. Arch Neurol. 67:688–696. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Mattsson N, Zetterberg H, Hansson O, Andreasen N, Parnetti L, Jonsson M, Herukka SK, van der Flier WM, Blankenstein MA, Ewers M, et al: CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. JAMA. 302:385–393. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Landau SM, Harvey D, Madison CM, Reiman EM, Foster NL, Aisen PS, Petersen RC, Shaw LM, Trojanowski JQ, Jack CR Jr, et al: Comparing predictors of conversion and decline in mild cognitive impairment. Neurology. 75:230–238. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Van Rossum IA, Vos SJ, Burns L, Knol DL, Scheltens P, Soininen H, Wahlund LO, Hampel H, Tsolaki M, Minthon L, et al: Injury markers predict time to dementia in subjects with MCI and amyloid pathology. Neurology. 79:1809–1816. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Vemuri P, Wiste HJ, Weigand SD, Shaw LM, Trojanowski JQ, Weiner MW, Knopman DS, Petersen RC and Jack CR Jr; Alzheimer's Disease Neuroimaging Initiative: MRI and CSF biomarkers in normal, MCI, and AD subjects: Predicting future clinical change. Neurology. 73:294–301. 2009. View Article : Google Scholar : PubMed/NCBI

55 

Eckerström C, Olsson E, Bjerke M, Malmgren H, Edman A, Wallin A and Nordlund A: A combination of neuropsychological, neuroimaging, and cerebrospinal fluid markers predicts conversion from mild cognitive impairment to dementia. J Alzheimers Dis. 36:421–431. 2013.PubMed/NCBI

56 

Shaffer JL, Petrella JR, Sheldon FC, Choudhury KR, Calhoun VD, Coleman RE and Doraiswamy PM; Alzheimer's Disease Neuroimaging Initiative: Predicting cognitive decline in subjects at risk for Alzheimer disease by using combined cerebrospinalfluid, MR imaging, and PET biomarkers. Radiology. 266:583–591. 2013. View Article : Google Scholar :

57 

Ewers M, Walsh C, Trojanowski JQ, Shaw LM, Petersen RC, Jack CR Jr, Feldman HH, Bokde AL, Alexander GE, Scheltens P, et al: Prediction of conversion from mild cognitive impairment to Alzheimer's disese dementia based upon biomarkers and neuropsychological test performance. Neurobiol Aging. 33:1203–1214. 2012. View Article : Google Scholar

58 

Vemuri P, Wiste HJ, Weigand SD, Knopman DS, Trojanowski JQ, Shaw LM, Bernstein MA, Aisen PS, Weiner M, Petersen RC, et al: Serial MRI and CSF biomarkers in normal aging, MCI, and AD. Neurology. 75:143–151. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Blennow K, Wallin A, Fredman P, Karlsson I, Gottfries CG and Svennerholm L: Blood-brain barrier disturbance in patients with Alzheimer's disease is related to vascular factors. Acta Neurol Scand. 81:323–326. 1990. View Article : Google Scholar : PubMed/NCBI

60 

Skoog I, Wallin A, Fredman P, Hesse C, Aevarsson O, Karlsson I, Gottfries CG and Blennow K: A population study on blood-brain barrier function in 85-year-olds: Relation to Alzheimer's disease and vascular dementia. Neurology. 50:966–971. 1998. View Article : Google Scholar : PubMed/NCBI

61 

Halliday MR, Pomara N, Sagare AP, Mack WJ, Frangione B and Zlokovic BV: Relationship between cyclophilin a levels and matrix metalloproteinase 9 activity in cerebrospinal fluid of cognitively normal apolipoprotein e4 carriers and blood-brain barrier breakdown. JAMA Neurol. 70:1198–1200. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Matsumoto Y, Yanase D, Noguchi-Shinohara M, Ono K, Yoshita M and Yamada M: Blood-brain barrier permeability correlates with medial temporal lobe atrophy but not with amyloid-beta protein transport across the blood-brain barrier in Alzheimer's disease. Dement Geriatr Cogn Disord. 23:241–245. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, Toga AW, Jacobs RE, Liu CY, Amezcua L, et al: Blood-brain barrier breakdown in the aging human hippo-campus. Neuron. 85:296–302. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Zetterberg H, Andreasson U, Hansson O, Wu G, Sankaranarayanan S, Andersson ME, Buchhave P, Londos E, Umek RM, Minthon L, et al: Elevated cerebrospinal fluid BACE1 activity in incipient Alzheimer disease. Arch Neurol. 65:1102–1107. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Ewers M, Zhong Z, Bürger K, Wallin A, Blennow K, Teipel SJ, Shen Y and Hampel H: Increased CSF-BACE 1 activity is associated with ApoE-epsilon 4 genotype in subjects with mild cognitive impairment and Alzheimer's disease. Brain. 131:1252–1258. 2008. View Article : Google Scholar : PubMed/NCBI

66 

Olsson A, Höglund K, Sjögren M, Andreasen N, Minthon L, Lannfelt L, Buerger K, Möller HJ, Hampel H, Davidsson P and Blennow K: Measurement of alpha- and beta-secretase cleaved amyloid precursor protein in cerebrospinal fluid from Alzheimer patients. Exp Neurol. 183:74–80. 2003. View Article : Google Scholar : PubMed/NCBI

67 

Perneczky R, Tsolakidou A, Arnold A, Diehl-Schmid J, Grimmer T, Förstl H, Kurz A and Alexopoulos P: CSF soluble amyloid precursor proteins in the diagnosis of incipient Alzheimer disease. Neurology. 77:35–38. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Lewczuk P, Popp J, Lelental N, Kölsch H, Maier W, Kornhuber J and Jessen F: Cerebrospinal fluid soluble amyloid-β protein precursor as a potential novel biomarkers of Alzheimer's disease. J Alzheimers Dis. 28:119–125. 2012.

69 

Hansson O, Zetterberg H, Buchhave P, Andreasson U, Londos E, Minthon L and Blennow K: Prediction of Alzheimer's disease using the CSF Abeta42/Abeta40 ratio in patients with mild cognitive impairment. Dement Geriatr Cogn Disord. 23:316–320. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Hölttä M, Hansson O, Andreasson U, Hertze J, Minthon L, Nägga K, Andreasen N, Zetterberg H and Blennow K: Evaluating amyloid-β oligomers in cerebrospinal fluid as a biomarker for Alzheimer's disease. PLoS One. 8:e663812013. View Article : Google Scholar

71 

Herskovits AZ, Locascio JJ, Peskind ER, Li G and Hyman BT: A Luminex assay detects amyloid β oligomers in Alzheimer's disease cerebrospinal fluid. PLoS One. 8:e678982013. View Article : Google Scholar

72 

Lesne SE, Sherman MA, Grant M, Kuskowski M, Schneider JA, Bennett DA and Ashe KH: Brain amyloid-β oligomers in ageing and Alzheimer's disease. Brain. 136:1383–1398. 2013. View Article : Google Scholar

73 

Savage MJ, Kalinina J, Wolfe A, Tugusheva K, Korn R, Cash-Mason T, Maxwell JW, Hatcher NG, Haugabook SJ, Wu G, et al: A sensitive aβ oligomer assay discriminates Alzheimer's and aged control cerebrospinal fluid. J Neurosci. 34:2884–2897. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Yang T, O'Malley TT, Kanmert D, Jerecic J, Zieske LR, Zetterberg H, Hyman BT, Walsh DM and Selkoe DJ: A highly sensitive novel immunoassay specifically detects low levels of soluble Aβ oligomers in human cerebrospinal fluid. Alzheimers Res Ther. 7:142015. View Article : Google Scholar

75 

Nabers A, Ollesch J, Schartner J, Kötting C, Genius J, Hafermann H, Klafki H, Gerwert K and Wiltfang J: Amyloid-β-secondary structure distribution in cerebrospinal fluid and blood measured by an immune-infrared-sensor: A biomarker candidate for Alzheimer's disease. Anal Chem. 88:2755–2762. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Kvartsberg H, Duits FH, Ingelsson M, Andreasen N, Öhrfelt A, Andersson K, Brinkmalm G, Lannfelt L, Minthon L, Hansson O, et al: Cerebrospinal fluid levels of the synaptic protein neurogranin correlates with cognitive decline in prodromal Alzheimer's disease. Alzheimers Dement. 11:1180–1190. 2015. View Article : Google Scholar

77 

Portelius E, Zetterberg H, Skillbäck T, Törnqvist U, Andreasson U, Trojanowski JQ, Weiner MW, Shaw LM, Mattsson N and Blennow K; Alzheimer's Disease Neuroimaging Initiative: Cerebrospinal fluid neurogranin: Relation to cognition and neurodegeneration in Alzheimer's disease. Brain. 138:3373–3385. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Kester MI, Teunissen CE, Crimmins DL, Herries EM, Ladenson JH, Scheltens P, van der Flier WM, Morris JC, Holtzman DM and Fagan AM: Neurogranin as a cerebrospinal fluid biomarker for synaptic loss in symptomatic Alzheimer disease. JAMA Neurol. 72:1275–1280. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Sjögren M, Rosengren L, Minthon L, Davidsson P, Blennow K and Wallin A: Cytoskeleton proteins in CSF distinguish frontotemporal dementia from AD. Neurology. 54:1960–1964. 2000. View Article : Google Scholar : PubMed/NCBI

80 

Sjögren M, Davidsson P, Gottfries J, Vanderstichele H, Edman A, Vanmechelen E, Wallin A and Blennow K: The cerebrospinal fluid levels of tau, growth-associated protein-43 and soluble amyloid precursor proteincorrelate in Alzheimer's disease, reflecting a common pathophysiological process. Dement Geriatr Cogn Disord. 12:257–264. 2001. View Article : Google Scholar

81 

de la Monte SM, Ghanbari K, Frey WH, Beheshti I, Averback P, Hauser SL, Ghanbari HA and Wands JR: Characterization of the AD7C-NTP cDNA expression in Alzheimer's disease and measurement of a 41-kDprotein in cerebrospinal fluid. J Clin Invest. 100:3093–3104. 1997. View Article : Google Scholar

82 

Tarawneh R, D'Angelo G, Macy E, Xiong C, Carter D, Cairns NJ, Fagan AM, Head D, Mintun MA, Ladenson JH, et al: Visinin-like protein-1: Diagnostic and prognostic biomarker in Alzheimer disease. Ann Neurol. 70:274–285. 2011. View Article : Google Scholar : PubMed/NCBI

83 

Tarawneh R, Lee JM, Ladenson JH, Morris JC and Holtzman DM: CSF VILIP-1 predicts rates of cognitive decline in early Alzheimer disease. Neurology. 78:709–719. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Tarawneh R, Head D, Allison S, Buckles V, Fagan AM, Ladenson JH, Morris JC and Holtzman DM: Cerebrospinal fluid markers of neurodegeneration and rates of brain atrophy in early Alzheimer disease. JAMA Neurol. 72:656–665. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Sutphen CL, Jasielec MS, Shah AR, Macy EM, Xiong C, Vlassenko AG, Benzinger TL, Stoops EE, Vanderstichele HM, Brix B, et al: Longitudinal cerebrospinal fluid biomarker changes in preclinical Alzheimer disease during middle age. JAMA Neurol. 72:1029–1042. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Czech C, Berndt P, Busch K, Schmitz O, Wiemer J, Most V, Hampel H, Kastler J and Senn H: Metabolite profiling of Alzheimer's disease cerebrospinal fluid. PLoS One. 7:e315012012. View Article : Google Scholar : PubMed/NCBI

87 

Liguori C, Stefani A, Sancesario G, Sancesario GM, Marciani MG and Pierantozzi M: CSF lactate levels, τ proteins, cognitive decline: A dynamic relationship in Alzheimer's disease. J Neurol Neurosurg Psychiatry. 86:655–659. 2015. View Article : Google Scholar

88 

Vafadar-Isfahani B, Ball G, Coveney C, Lemetre C, Boocock D, Minthon L, Hansson O, Miles AK, Janciauskiene SM, Warden D, et al: Identification of SPARC-like 1 protein as part of a biomarker panel for Alzheimer's disease in cerebrospinal fluid. J Alzheimers Dis. 28:625–636. 2012.

89 

Wildsmith KR, Schauer SP, Smith AM, Arnott D, Zhu Y, Haznedar J, Kaur S, Mathews WR and Honigberg LA: Identification of longitudinally dynamic biomarkers in Alzheimer's disease cerebrospinal fluid by targeted proteomics. Mol Neurodegener. 9:222014. View Article : Google Scholar : PubMed/NCBI

90 

Korff A, Liu C, Ginghina C, Shi M and Zhang J; Alzheimer's Disease Neuroimaging Initiative: α-Synuclein in cerebrospinal fluid of Alzheimer's disease and mild cognitive impairment. J Alzheimers Dis. 36:679–688. 2013.

91 

Olsson B, Hertze J, Ohlsson M, Nägga K, Höglund K, Basun H, Annas P, Lannfelt L, Andreasen N, Minthon L, et al: Cerebrospinal fluid levels of heart fatty acid binding protein are elevated prodromally in Alzheimer's disease and vascular dementia. J Alzheimers Dis. 34:673–679. 2013.

92 

Yamagishi S, Inagaki Y, Takeuchi M and Sasaki N: Is pigment epithelium-derived factor level in cerebrospinal fluid a promising biomarker for early diagnosis of Alzheimer's disease? Med Hypotheses. 63:115–117. 2004. View Article : Google Scholar : PubMed/NCBI

93 

Lovell MA, Lynn BC, Xiong S, Quinn JF, Kaye J and Markesbery WR: An aberrant protein complex in CSF as a biomarker of Alzheimer disease. Neurology. 70:2212–2218. 2008. View Article : Google Scholar : PubMed/NCBI

94 

Brys M, Pirraglia E, Rich K, Rolstad S, Mosconi L, Switalski R, Glodzik-Sobanska L, De Santi S, Zinkowski R, Mehta P, et al: Prediction and longitudinal study of CSF biomarkers in mild cognitive impairment. Neurobiol Aging. 30:682–690. 2009. View Article : Google Scholar :

95 

Ayton S, Faux NG and Bush AI; Alzheimer's Disease Neuroimaging Initiative: Ferritin levels in the cerebrospinal fluid predict Alzheimer's disease outcomes and are regulated by APOE. Nat Commun. 6:67602015. View Article : Google Scholar : PubMed/NCBI

96 

Ghanbari H, Ghanbari K, Beheshti I, Munzar M, Vasauskas A and Averback P: Biochemical assay for AD7C-NTP in urine as an Alzheimer's disease marker. J Clin Lab Anal. 12:285–288. 1998. View Article : Google Scholar : PubMed/NCBI

97 

Goodman I, Golden G, Flitman S, Xie K, McConville M, Levy S, Zimmerman E, Lebedeva Z, Richter R, Minagar A and Averback P: A multi-center blinded prospective study of urine neural thread protein measurements in patients with suspected Alzheimer's disease. J Am Med Dir Assoc. 8:21–30. 2007. View Article : Google Scholar : PubMed/NCBI

98 

de la Monte SM and Wands JR: The AD7c-ntp neuronal thread protein biomarker for detecting Alzheimer's disease. Front Biosci. 7:d989–d996. 2002.PubMed/NCBI

99 

Ma L, Chen J, Wang R, Han Y, Zhang J, Dong W, Zhang X, Wu Y and Zhao Z: The level of Alzheimer-associated neuronal thread protein in urine may be an important biomarker of mild cognitive impairment. J Clin Neurosci. 22:649–652. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Zengi O, Karakas A, Ergun U, Senes M, Inan L and Yucel D: Urinary 8-hydroxy-2′-deoxyguanosine level and plasma paraoxonase 1 activity withAlzheimer's disease. Clin Chem Lab Med. 50:529–534. 2011.PubMed/NCBI

101 

Kim KM, Jung BH, Paeng KJ, Kim I and Chung BC: Increased urinary F(2) isoprostanes levels in the patients with Alzheimer's disease. Brain Res Bull. 64:47–51. 2004. View Article : Google Scholar : PubMed/NCBI

102 

Yoshida M, Higashi K, Kuni K, Mizoi M, Saiki R, Nakamura M, Waragai M, Uemura K, Toida T, Kashiwagi K and Igarashi K: Distinguishing mild cognitive impairment from Alzheimer's disease with acrolein metabolites and creatinine in urine. Clin Chim Acta. 441:115–121. 2015. View Article : Google Scholar

103 

Rabassa M, Cherubini A, Zamora-Ros R, Urpi-Sarda M, Bandinelli S, Ferrucci L and Andres-Lacueva C: Low levels of a urinary biomarker of dietary polyphenol are associated with substantial cognitive decline over a 3-year period in older adults: The invecchiare in chianti study. J Am Geriatr Soc. 63:938–946. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Cook CN, Murray ME and Petrucelli L: Understanding biomarkers of neurodegeneration: Novel approaches to detecting tau pathology. Nat Med. 21:219–220. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Kfoury N, Holmes BB, Jiang H, Holtzman DM and Diamond MI: Trans-cellular propagation of Tau aggregation by fibrillar species. J Biol Chem. 287:19440–19451. 2012. View Article : Google Scholar : PubMed/NCBI

106 

Holmes BB, Furman JL, Mahan TE, Yamasaki TR, Mirbaha H, Eades WC, Belaygorod L, Cairns NJ, Holtzman DM and Diamond MI: Proteopathic tau seeding predicts tauopathy in vivo. Proc Natl Acad Sci USA. 111:E4376–E4385. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Yanamandra K, Kfoury N, Jiang H, Mahan TE, Ma S, Maloney SE, Wozniak DF, Diamond MI and Holtzman DM: Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron. 80:402–414. 2013. View Article : Google Scholar : PubMed/NCBI

108 

Kong D, Giovanello KS, Wang Y, Lin W, Lee E, Fan Y, Murali Doraiswamy P and Zhu H: Predicting Alzheimer's disease using combined imaging-whole genome SNP data. J Alzheimers Dis. 46:695–702. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Sun Y, Bresell A, Rantalainen M, Höglund K, Lebouvier T and Salter H; Alzheimer Disease Neuroimaging Initiative: An integrated bioinformatics approach for identifying genetic markers that predict cerebrospinal fluid biomarker p-tau181/Aβ1-42 ratio in ApoE4-negative mild cognitive impairment patients. J Alzheimers Dis. 45:1061–1076. 2015.

110 

Castro-Chavira SA, Fernandez T, Nicolini H, Diaz-Cintra S and Prado-Alcala RA: Genetic markers in biological fluids for aging-related major neurocognitive disorder. Curr Alzheimer Res. 12:200–209. 2015. View Article : Google Scholar : PubMed/NCBI

111 

Femminella GD, Ferrara N and Rengo G: The emerging role of microRNAs in Alzheimer's disease. Front Physiol. 6:402015. View Article : Google Scholar : PubMed/NCBI

112 

Namioka N, Hanyu H, Hirose D, Hatanaka H, Sato T and Shimizu S: Oxidative stress and inflammation are associated with physical frailty in patients with Alzheimer's disease. Geriatr Gerontol Int. Jun 14–2016.Epub ahead of print. View Article : Google Scholar

113 

Panza F, Solfrizzi V, Barulli MR, Santamato A, Seripa D, Pilotto A and Logroscino G: Cognitive Frailty: A systematic review of epidemiological and neurobiological evidence of an age-related clinical condition. Rejuvenation Res. 18:389–412. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 14 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ruan Q, D'Onofrio G, Sancarlo D, Greco A and Yu Z: Potential fluid biomarkers for pathological brain changes in Alzheimer's disease: Implication for the screening of cognitive frailty. Mol Med Rep 14: 3184-3198, 2016
APA
Ruan, Q., D'Onofrio, G., Sancarlo, D., Greco, A., & Yu, Z. (2016). Potential fluid biomarkers for pathological brain changes in Alzheimer's disease: Implication for the screening of cognitive frailty. Molecular Medicine Reports, 14, 3184-3198. https://doi.org/10.3892/mmr.2016.5618
MLA
Ruan, Q., D'Onofrio, G., Sancarlo, D., Greco, A., Yu, Z."Potential fluid biomarkers for pathological brain changes in Alzheimer's disease: Implication for the screening of cognitive frailty". Molecular Medicine Reports 14.4 (2016): 3184-3198.
Chicago
Ruan, Q., D'Onofrio, G., Sancarlo, D., Greco, A., Yu, Z."Potential fluid biomarkers for pathological brain changes in Alzheimer's disease: Implication for the screening of cognitive frailty". Molecular Medicine Reports 14, no. 4 (2016): 3184-3198. https://doi.org/10.3892/mmr.2016.5618