Next Article in Journal
Influence of SH2B3, MTHFD1L, GGCX, and ITGB3 Gene Polymorphisms on theVariability on Warfarin Dosage Requirements and Susceptibility to CVD in the Jordanian Population
Next Article in Special Issue
Biomarkers for Alzheimer’s Disease (AD) and the Application of Precision Medicine
Previous Article in Journal / Special Issue
The Other Side of Alzheimer’s Disease: Influence of Metabolic Disorder Features for Novel Diagnostic Biomarkers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer’s Disease: Sensitivity, Specificity and Potential for Clinical Use

by
Cristina d’Abramo
1,
Luciano D’Adamio
2 and
Luca Giliberto
1,3,4,*
1
The Litwin-Zucker Center for Alzheimer’s Disease & Memory Disorders, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA
2
Rutgers Alzheimer’s Disease & Dementia Research Center (RUADRC), Rutgers Brain Health Institute, Rutgers University, Piscataway, NJ 08854, USA
3
Northwell Health Neuroscience Institute, Northwell Health System, Manhasset, NY 11030, USA
4
Donald and Barbara Zucker School of Medicine, Hofstra University, Hempstead, NY 11549, USA
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2020, 10(3), 116; https://doi.org/10.3390/jpm10030116
Submission received: 1 August 2020 / Revised: 21 August 2020 / Accepted: 1 September 2020 / Published: 8 September 2020
(This article belongs to the Special Issue Novel Biomarkers in Alzheimer’s Disease)

Abstract

:
Alzheimer’s disease (AD) is the most common type of dementia, affecting more than 5 million Americans, with steadily increasing mortality and incredible socio-economic burden. Not only have therapeutic efforts so far failed to reach significant efficacy, but the real pathogenesis of the disease is still obscure. The current theories are based on pathological findings of amyloid plaques and tau neurofibrillary tangles that accumulate in the brain parenchyma of affected patients. These findings have defined, together with the extensive neurodegeneration, the diagnostic criteria of the disease. The ability to detect changes in the levels of amyloid and tau in cerebrospinal fluid (CSF) first, and more recently in blood, has allowed us to use these biomarkers for the specific in-vivo diagnosis of AD in humans. Furthermore, other pathological elements of AD, such as the loss of neurons, inflammation and metabolic derangement, have translated to the definition of other CSF and blood biomarkers, which are not specific of the disease but, when combined with amyloid and tau, correlate with the progression from mild cognitive impairment to AD dementia, or identify patients who will develop AD pathology. In this review, we discuss the role of current and hypothetical biomarkers of Alzheimer’s disease, their specificity, and the caveats of current high-sensitivity platforms for their peripheral detection.

1. Introduction

1.1. Clinicopathological Definition of Alzheimer’s Disease

Alzheimer’s disease (AD) is a neurodegenerative disorder, characterized by progressive cognitive impairment, affecting the domains of memory, speech, praxis, awareness and executive function, representing a decline from the prior functional state, with a relentless course [1]. It is the most common cause of dementia. Along with the destructive nature of the disease, which affects patients and their families, AD represents an enormous burden on public health. An estimated of 5+ million Americans have AD, and the prevalence worldwide reaches almost 60 million; the diseases hits the elderly population the most, with 11% of individuals 65 and older affected by the disease [2,3]. Two thirds of AD patients are women, implying hormonal, lifestyle and genetic factors as co-causative elements [2,4,5,6,7,8,9,10,11,12,13,14,15]. AD is the sixth leading cause of death in the US [16] and mortality due to AD has steadily increased in the last 20 years, in contrast to opposite trends for other chronic diseases, such as cardiovascular, cancer, HIV and more. The socio-economic burden of the disease, on patients, families and caregivers at large, and on society, is unparalleled; AD is the disease that contributes most to disability and poor health in the US, reducing life expectancy and productivity of the affected individuals and the wellbeing of caregivers, who often have to give up jobs and careers to care for their loved ones [17,18,19]; this is often disproportionally affecting women too [20,21,22]. Finally, AD has a tremendous economic impact: the sole health care costs, variably covered by insurances (including Medicare and Medicaid) or out of pocket, to care for this disease, is in excess of 230 billion [23]. Age-matched comparisons show that individuals affected by dementia had 3–4x the annual health care expenses than those not affected. It is estimated that these costs will reach, in 2050, 1 trillion dollars (2020 Alzheimer’s Disease Facts and Figures. Available online: https://www.alz.org/media/Documents/alzheimers-facts-and-figures.pdf. Accessed on 29 July 2020), a figure comparable to the US budget deficit in 2020. Interestingly, these numbers should probably be corrected upward, as the disease is often under-recognized and under-reported. This is particularly relevant to our discussion, given current limitations in performing early and accurate diagnosis.
The pathological diagnosis of AD relies on the assessment of specific brain features [24,25,26,27], based on the presence, morphology and density of lesions and their topographic distribution, in the appropriate clinical setting. Alzheimer’s brains are characterized by a spatiotemporally defined accumulation of amyloid beta (Aβ) plaques, neurofibrillary tangles (NFT), of hyperphosphorylated and aggregated tau and neuronal loss [25,28,29]. Aβ accumulates mostly as the Aβ peptide ending at residue 42, which is considered the most toxic and most prone to aggregation [30]; tau accumulates as a hyperphosphorylated and conformationally altered form, in both its 3- and 4-repeats variants [31]. The trajectory of protein accumulation is relevant. It is speculated that Aβ accumulates first; this is followed by the hyperphosphorylation, conformational changes and aggregation of tau; finally, neuronal loss ensues.
The link between these three features is, to some degree, still loose. Both Aβ [30,32] and tau [33,34] are per se neurotoxic; Aβ accumulation can trigger tau accumulation [35] but, in several models, tau can independently aggregate and cause neurodegeneration [36]. Furthermore, amyloid deposition, by pathological and imaging staging (amyloid positron emission tomography—PET), does not topographically and functionally correlate with the clinical cognitive decline seen in our patients, while tau does, and there are cases of aged individuals who have amyloid deposition in their brain, but no tau and no clinical dementia [25,37,38].
A significant degree of co-pathology, not strictly specific of AD, is common in these brains [39]. This includes the presence of activated microglia and reactive astrocytes, which appear initially around neuritic plaques, and increase in density and activation in proportion to the neuronal damage and the presence of tau pathology [40,41,42]; vascular alterations, in the form of amyloid deposition in the blood vessels (CAA: cerebral amyloid angiopathy); accumulation of other structural proteins in neurons, such as actin and actin-associated proteins in the form of Hirano bodies [43]; granulovacuolar degeneration (GVD), a feature often associated with the initial hyperphosphorylation and aggregation of tau in the hippocampus [44]. Argyrophilic grains of 4-repeat tau can also be found in neuronal dendrites, near oligodendrocytes inclusions and seems to be relevant to disease progression [45].
In some AD cases, specific neuronal populations are vulnerable to develop Lewy body pathology of α-synuclein [43,46], which is a signature of Parkinson’s disease (PD), mostly at the axonal level, in cortico-limbic regions, olfactory bulbs and substantia nigra. Finally, TAR DNA-binding protein 43 (TDP-43) pathology, more commonly found in cases of frontotemporal lobe degeneration (FTD), can be present in AD, in the amygdala, hippocampus and eventually in the neocortex and more diffusely [47,48], being not specific of AD, but underlying the vast array of cellular dysfunction present in this disease, which can affect RNA-related metabolism as well [49]. The common path, which is likely consequent to all the above changes, is the dramatic loss of synapses, which precedes neuronal loss, and mostly follows the distribution of tau pathology [43,50,51,52].
The routine clinical diagnosis of possible or probable AD is currently based on the identification of symptoms by the clinician, mostly Neurologists, Geriatricians, Psychiatrists and Primary Care Physicians, followed by more in-depth assessment by Neuropsychology testing and using the tools available in clinical practice, e.g., Magnetic Resonance Imaging (MRI) of the brain and common blood works. Once AD is suspected, these tools allow to rule out secondary dementia and other, different pattern of primary dementia. On a more sophisticated level, the analysis of the cerebrospinal fluid (CSF), the use of PET imaging and genetics (in familial cases) may allow us to obtain a definitive diagnosis of AD in vivo: due to cost and perception of invasiveness, this is often limited to Academic centers or in the context of clinical trials.
Ideally, we should diagnose AD, and every dementia, as early as possible. Mild cognitive impairment (MCI) is a prodromal phase of dementia, where the patient shows symptoms of cognitive changes but maintains good functionality. This is a moment where the application of biomarkers is paramount and significant therapeutic intervention may still possible. Unfortunately, there is a significant underappreciation of the disease, which is missed in more than 20% of the cases. On the other hand, lack of familiarity with alternative diagnoses, leads to overdiagnosis of AD in more than 25% of the cases, where potentially curable conditions are instead the underlying cause of the cognitive and functional decline [53,54,55,56].

1.2. Pathogenesis

The majority of our understanding of the pathogenesis of AD derives from the identification of key genes involved in the etiology and pathology of the disease. In a small percentage of the cases, the disease is inherited as autosomal dominant (FAD: familial AD), with an early age of onset, as early as the ‘40s, and a faster progression [57]. FAD is caused by mutations of one of three genes, Amyloid Precursor Protein (APP), Presenilin 1 (PS1) or Presenilin 2 (PS2). APP is the precursor of the amyloid peptide, Aβ, which is generated from APP by two sequential cleavages: first, the beta-secretase activity of BACE1 (beta-site amyloid precursor protein cleaving enzyme 1) generates a peptide of ~99 amino acids (C99); subsequently, the activity of the gamma-secretase, a multi-peptide protease, progressively cleaves C99 to generate the Aβ peptide ending at residues 40 or 42. The gamma secretase complex is composed of PS1 or PS2 and three other proteins, nicastrin, Aph1 and Pen2 [58]. Mutations in APP mostly lead to increase affinity of APP itself for BACE1, with increased generation of C99. Mutations of PS1 and PS2, which result in an actual loss of function mutations of gamma-secretase [59,60,61,62], lead to increased generation of the Aβ peptide ending in residue 42. This is recognized as the most toxic of the Aβ peptides [63]. About 95% of AD cases are sporadic (SAD) and have a later onset (after 65 years of age): although SAD cases lack a Mendelian cause, variants of genes such as Apolipoprotein E (APOE) [64,65], and triggering receptors expressed on myeloid cells 2 (TREM2) [66] are known to increase the risk of developing SAD. It is not clear what the initial trigger to Aβ and tau accumulation is in the sporadic disease. At the subcellular and molecular level, the complexity of Aβ and tau accumulation and their specific contribution to pathology has been extensively studied, and is beyond the scope of the current publication. The general understanding is that both amyloid and tau oligomers have the ability to disrupt several of the essential functions of neurons, and possibly other brain cells, i.e., metabolic and mitochondrial activity [67,68], synaptic communication [69,70], membrane integrity [71], membrane pore formation with ions leakage [72,73], protein trafficking and degradation [74,75]. Both Aβ and tau possess the ability to trigger a cascade of functional changes in neurons, microglia, astrocytes and oligodendrocytes, which results in neuronal dysfunction, synaptic loss, innate immune system activation, inflammation, blood-brain barrier impairment and neurodegeneration [39,76]. Relevant to our discussion, the immune system is activated (early) in AD, and may contribute, by means of ineffective clearance of Aβ and tau by the microglia, to the diffusion of such pathology to neighboring or connected areas [77,78], and inflammation per se is a trigger to further production of Aβ and activation of kinases to hyper-phosphorylate tau [79], cascading more neuropathology.

1.3. The (Amyloid, Tau, Neurodegeneration) ATN System

Stemming from the above definitions, pathological correlates and pathogenic theories, AD can be viewed as the combination of specific brain pathology, Aβ and tau, and neurodegeneration, which eventually results in the clinical presentation. Recent advances in brain imaging and more sensitive and specific means to detect Aβ and tau in biological fluids, associated with our ability to detect loss of brain matter, metabolism or function, have allowed for development of a classification and staging system for AD, the ATN system [80], which is routinely revised [81], to include the everchanging horizon of new biomarkers available. Patients are classified by positivity to markers of amyloid deposition (A), tau load (T) and neurodegeneration (N). Amyloid markers include the positivity to Amyloid PET (several tracers are now available) or reduced levels of cerebrospinal fluid (CSF) Aβ42, Aβ40 or of the Aβ42/40 ratio (see below). T is defined by the high levels of total or phosphorylated tau in the CSF (see below), although the new available tau PET tracers may soon be included in the criteria. Finally, N is related to the neurodegenerative aspect, and can be defined by loss of volume/activity in specific areas of the brain by magnetic resonance imaging (MRI, e.g., NeuroQuant, [82,83]), fluorodeoxyglucose (FDG)-PET or by the presence of CSF biomarkers of synaptic or neuronal loss, e.g., Neurofilament Light chain (NfL).
This system has been a useful tool for many biomarkers studies, both in biological fluids and imaging, remaining the “benchmark” for AD diagnosis and prognosis determination. Importantly, the ATN system can be extremely useful to identify and correctly classify individuals who may have amyloid deposition but no neurodegeneration (A+T-N-) and vice versa (A-T-N+); this is extremely important when studying pathophysiological correlates and in longitudinal assessments. As hinted above, a good proportion of the ageing population will show deposition of amyloid in their brain, but no clinical dementia; we know that this amyloid may not be the same amyloid that accumulates in AD proper [84], hence not cause the disease, but also other factors may not be present in these individuals that predispose to tau accumulation and neurodegeneration. On the other hand, neurodegeneration and dementia may occur because of pathology other than Alzheimer’s, e.g., vascular, TDP43, alpha-synuclein, etc. If rigorously determined and applied to our studies, the ATN system will be invaluable, as it prescinds from the subjective interpretation of the clinical status of the patients, and is purely descriptive of their biomarkers status.
It must be noted, though, that not all the pathophysiological events and biomarkers in AD can be inscribed in the ATN definition, hence we will be presenting a wider paradigm, that includes, for example, inflammation.

1.4. Treatment Options

No disease-modifying treatment is available as of today, despite decades of efforts aimed to curb the pathological changes of the disease [85,86,87]. The widest attempts to tackle the disease have been directed against the production/accumulation of Aβ and have overall been underwhelming [85]. Reducing its production via inhibition of BACE or gamma-secretase has not been effective, resulting in potential side effects, included hastening the cognitive decline [86]; active vaccination efforts against Aβ has also not been successful, mostly due to cases of dramatic adverse events, in the form of hemorrhagic encephalitis [88], despite some cognitive benefits in the survivors [89]. Alternative active vaccinations strategies, aimed at inducing a wide reactivity to Aβ without the excessive inflammatory response, have been tested [90,91] with scarce success. Passive immunization against Aβ was also tainted by questionable clinical efficacy and concerns of serious side effects [92,93,94,95], but efforts are well under way: at the moment that we write this review, a monoclonal antibody against amyloid has been submitted for U.S. Food and Drug Administration (FDA) expedited approval as a modifying drug against AD, with some skepticism among the scientific community in respect to the available efficacy data [95] (NCT02477800). Tau has been the other logical therapeutic target. Current strategies are directed to target total tau, which is thought to accumulate intra- and extracellularly, and may spread from cell to cell, aggregating and getting hyperphosphorylated, contributing to neurodegeneration. Antibodies targeting extracellular tau have been in clinical trial now for some time, and we are all bracing for the upcoming results. Of note, this strategy has so far failed in progressive supranuclear palsy (PSP) cases with tau pathology (NCT03068468). An active immunization approach against tau is currently under way as well [96,97].
What clinicians are left with is the use of symptomatic treatments. Acetylcholine deficits are well recognized in AD [98], and are the basis for acetylcholine esterase inhibitors, which can ameliorate quality of life. Memantine, a noncompetitive N-methyl-D-aspartate-receptor antagonist, has also shown some benefit in maintaining cognitive function, alone and in association with acetylcholine esterase inhibitors. In all, unfortunately, the enthusiasm for these agents is very low, due to low efficacy and possible adverse events, such as behavioral changes, heart arrhythmias, loss of appetite and weight, which in the elderly population are a major concern.
The neuropathological changes described above are projected to begin decades before the presentation of symptoms. This makes any medical intervention, even in the prodromal stages, a “too little-too late” approach, with few chances of being meaningful. The challenges in AD are, in our opinion, 2-fold; first, defining the actual pathogenesis of the disease, by far not yet clear; second, be able to identify the risk to develop the disease early enough so that disease-modifying intervention can be effective.

1.5. Definition of AD Biomarkers and Diagnostic Challenges

What is a biomarker? What is an adequate and useful diagnostic biomarker for Alzheimer’s disease? While it is fairly easy to respond to the first question, it is very difficult to answer the second. The reason for the latter is that, save for the genetically determined cases of AD (a minority of the cases), in the absence of a univocal and comprehensive pathogenic pathway, it is unlikely to be able to diagnose AD with certainty in the living adult in a time frame that would be useful for significant intervention.
A biomarker is a “defined characteristic that is measured as an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention, including therapeutic interventions” (FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools). Available online: https://www.ncbi.nlm.nih.gov/books/NBK326791/. Accessed on 29 July 2020). This definition includes both a diagnostic and prognostic element. The current literature about AD biomarkers is extensive and rich of analyses and reviews. The popular website, Alzforum, has its own dedicated meta-analysis page (Alzforum. Available online: https://www.alzforum.org/alzbiomarker. Accessed on 29 July 2020).
AD biomarkers are quite reliable indicators of disease presence, topographical extension and stage. Levels of Aβ, total tau (t-tau), and phosphorylated tau (p-tau) are concurrent with AD pathology [99,100] and indicate its presence; at this stage amyloid [101] and tau PET imaging [102] define the topographical extension of such pathology, while MRI [103] and FDG-PET [104] define the topographical severity of the neurodegenerative process; finally, markers such as p-tau, NfL [105] and YKL40 [106] can be useful, and early, peripheral indicators of disease stage.
We can use a combination of fluid and imaging biomarkers to complement the clinical investigation in our patients, and fulfill the current diagnostic criteria [107,108,109,110] with ease. One caveat is that while these criteria can work well in research settings [111,112], the reality of clinical practice is tainted by the reluctance of patients to have invasive testing done (CSF) and by the lack or insurance support for the more “fancy” imaging [113]. Fortunately, as described below, the rapid evolution of detection techniques is allowing to reach remarkable results from blood biomarkers, which would improve access for patients.
Another element must be taken in consideration when defining a useful biomarker: time. The concept of early diagnosis has become crucial in neurodegeneration, since protein aggregation and accumulation might only reflect the last stage of the pathogenic cascade, a point of no return in the clinical manifestation and progression of the diseases, where therapeutic interventions have so far been ineffective. As mentioned above, the exact succession of pathogenic events in AD is still unclear; Aβ and tau may be causative, but subtler prior changes in neuronal connectivity, metabolic patterns and inflammation could represent an early disruption of neuronal function, leading to secondary accumulation of Aβ and tau [36]. In this context, an early diagnosis should go beyond Aβ and tau. A “late” biomarker’ profile may also be tainted by ongoing and long-standing pathology, and by other factors related to ageing, e.g., brain trauma, infections, microvascular disease/strokes, and co-morbidity with other frequent neurodegenerative conditions such as PD [114]. The clinical presentation of FAD and SAD is similar, except for the age of onset. For FAD mutations carriers, preclinical diagnosis is possible by genetic analysis; the more common SAD cases, instead, remain elusive until clinically evident. Hence, the challenge of procuring an early preclinical diagnosis of disease or susceptibility to it.
With these limitations in mind, the science of AD biomarkers has evolved tremendously in the last decade, and we are looking now at the possibility to have reliable peripheral non-invasive, scalable and affordable biomarkers of AD pathology and disease stage.
We will review below the current, most promising fluid (blood and CSF) biomarkers available, discuss the pathophysiologic correlate of each, their clinical significance, their general applicability in the clinical and research setting. We will then take tau as an example of biomarker detection complexity, with blessings and caveats, stemming from the need to accurately validate data emerging from new ultra-sensitive platforms, in light of their biochemical limitations.
Importantly, we have divided the below presentation in biomarkers that are specific of AD, where their appropriate combination in biological fluids defines the disease, from biomarkers that are representative of neurodegeneration or associated pathology, such as synaptic and neuronal loss, inflammation, vascular disease and others. The latter biomarkers may be present in neurodegenerative diseases different from AD, in head trauma, in stroke and infections, hence they are not specific for AD. They are, though, extremely useful to follow, and at time predict, AD progression and presentation. Their precise causative role in the neurodegenerative process is still unknown, hence we will present the current known hypotheses in descriptive fashion.

2. Biomarkers of AD Pathology

2.1. Aβ

Given its pathophysiological role, Aβ has revealed itself as the major biomarker of the disease, in addition to being one of main therapeutic targets so far. Over the last 30+ years, exponential efforts have demonstrated that CSF (and more recently blood) levels of Aβ (1) are reduced in AD compared to non-demented controls [115], (2) are decreased in mild cognitive impairment (MCI) patients compared to normal controls [116], (3) the trend of Aβ decrease is predictive of the clinical progression of MCI to AD and preclinical AD [117,118] to MCI. Similarly, and (4) the Aβ42/40 ratio is reduced in MCI and AD, with better clinical correlations to than total Aβ [119,120].
What is the pathogenic significance of Aβ reduction in biological fluids? Why is the Aβ42/40 ratio going down as the disease progresses? Is there ever a time, in the preclinical phases of the disease, when Aβ is instead increased in biological fluids, possibly marking a pivotal moment where a therapeutic intervention would still be effective?
In FAD, APP mutations lead to an increased overall production of Aβ, since the amyloidogenic cleavage of APP is favored. Mutations in PS1 or PS2 result instead in a preferential cleavage of C99 to form the Aβ42, while in reality the overall amount of Aβ may be reduced, since these are loss of function mutations [121]. In either case, there is an absolute or relative abundance of Aβ42 released in the brain parenchyma, which has a high tendency to aggregate and deposit as oligomers, fibrils and plaques in a process of nucleation [122]. Furthermore, BACE cleavage is seldom limited to residue #1 of Aβ; the amyloid peptide often starts at residue #3 or #11, a glutamate residue (E), which tends to be cyclized [123,124] as pyro-glutamate (pE). The resulting AβN3(pE) and AβN11(pE) peptides also have high tendency to aggregation, adding to the stickiness of Aβ42, and have been considered one of the first seeding species of Aβ [125]. In this context, it is easy to understand how the parenchymal sequestration of Aβ results in lower level of the soluble molecule available to diffuse to the interstitial fluid, CSF and blood, hence the low levels thereof. Further decrease of Aβ in the periphery may also be related to the progressive and massive loss of neuronal cells along the progression of the disease, as neurons are the highest manufacturers of the peptide [126]. Finally, other factors such as inflammation, infection or perturbation of Aβ clearance mechanism may affect its peripheral levels, though unlikely to have a chronic effect [127].
Other conditions show the opposite; in traumatic brain injury (TBI), for example, CSF and plasma levels of Aβ1-42 increase steadily after the acute event, and normalize with recovery in weeks [127,128,129]; in some cases, an increase of APP itself was noted when sampling intraventricular CSF [130]. These data tend to correlate with other markers of neuronal damage, i.e., the release of total tau, neuronal specific enolase (NSE) and APOE, but the actual mechanism of shedding is unclear. Notably, axonal injury after TBI is associated with redistribution of APP towards the axons and increased processing to generate Aβ, explaining part of the CSF levels [131,132].
Interestingly, whereas Aβ accumulation in the brain results in a reduction in the periphery, tau accumulation in the brain parallels its peripheral abundance [115] (discussed below). Tau accumulation follows, in time, Aβ accumulation. Can this time difference (maybe decades) explain the dichotomy? If so, is there a moment in time in AD pathogenesis when Aβ levels are actually increased or normal in the periphery? The current data on autosomal dominant AD show initial amyloid-PET abnormalities ca. 22 years prior the expected symptom onset (EYO-22) and CSF Aβ reduction (and tau increase) a few years later (EYO-19 to -14) [133]. This series of events signifies that (1) sensible Aβ deposition in the brain happens quite early in the pathogenesis, (2) current CSF (and blood) biomarkers follow brain changes by several years, and (3) measures of neuronal damage (peripheral tau, etc.) do not fall far behind peripheral Aβ changes. This leaves open the question of what happens even earlier to peripheral Aβ, is it ever elevated? Is that the ideal moment where anti-Aβ therapeutic strategies would be most effective? Only longitudinal studies with a very early CSF and blood sampling will be able to answer these questions.
A recent study in a Down syndrome (DS) cohort [134], with CSF sampling from age 30 to 61, showed Aβ42 reductions which was proportional to age and cognitive decline (from cognitive stable, to MCI to AD), with higher levels than those reported in corresponding older non-DS controls, MCI and AD [135], but did not have a non-DS age-matched control group. In another study [136], CSF levels of Aβ peptide were found to be elevated from 8 to 54 months of age in DS children, while tau was low, likely due to Aβ overproduction in the absence of significant parenchymal sequestration and neuronal loss at that age.
CSF. Sampling CSF by lumbar puncture (LP) is the most direct way to obtain biomarkers that reflect what happens in the brain. Being invasive, most patients do not feel comfortable undergoing a spinal tap, although the rate of complications, i.e., post-LP headache, bleeds or infections is very low [137]. CSF levels and variations of Aβ have long been the hallmark features of AD-type dementia. The literature on the matter is extremely extensive and will not be reviewed here. We will limit our discussion to the clinicopathological relevance of Aβ in CSF. The first attempts at measuring soluble APP-derived fragment in the CSF of AD patients were carried out by D. Selkoe and S. Younkin [138] and B. Frangione [139] labs, determining the reduction of soluble APP (sAPP) fragments in the CSF of AD patients. Soon after, scientists were able to map the Aβ region and retrieve the peptide in the CSF [140]. This led to the first studies looking at differences between AD and control subjects in soluble APP and Aβ peptides [141,142,143,144,145], with notable differences likely due to detection methods and populations [146,147].
CSF levels of Aβ 42 are generally lower in AD compared to control subjects, and have been seen to decline with disease progression [135,147,148,149,150]. Aβ1-42 levels can be reduced as much as 50% in patients with AD compared to control subjects [151]. There seems to be, though, significant overlap of total Aβ1-42 levels between AD and non-AD dementia, such as vascular dementia, Parkinson’s disease dementia (PDD), Lewy body disease (LBD), frontotemporal lobe degeneration (FTD) and even amyotrophic lateral sclerosis (ALS), and this may be due to population selection, stage of disease or overlap of pathogenic events in advanced disease [152,153,154,155,156,157,158].
In the pre-dementia stages, both amnestic MCI (aMCI), the most frequent precursor of AD, and non-amnestic MCI, showed low levels of Aβ1-42 [154,159,160,161,162] compared to control subjects, also with significant overlap.
In all, the use of Aβ1-42 alone does not seem to be too promising for differentiating AD from non-AD dementia and less so at the MCI stage, due to heterogeneity and overlap. As mentioned above, the pathophysiology of APP processing in AD is characterized by an imbalance of the 1-42 over the 1-40 species of Aβ, with preferential deposition of the former in the brain. Additionally, the gamma-secretase complex cleaves APP in a sequential fashion, generating progressively shorter fragments, like the Aβ1-38. These fragments have been identified in CSF, and may be useful biomarkers as well [162]. Thus, measuring the CSF ratio of Aβ42/40 (and other 42/X peptides ratios, e.g., 42/38, etc.) has been attempted. Indeed, the CSF ratio of Aβ42/40 has proven to be a reliable marker of AD, both at the dementia and MCI stage, as well as of the risk of progression from MCI to AD [163,164,165,166]. Furthermore, this ratio was also useful to differentiate AD from other non-AD cognitive changes, such as subcortical deficits of vascular disease. Such correlations were validated by both amyloid PET and extensive clinical and conventional MRI approaches. CSF Aβ42/40 correlates almost completely with amyloid PET, and when there is discordance, longitudinal studies have shown that PET imaging turns positive within a few years [117,119,150]. Finally, it was shown that different assay platforms resulted in similar results [167].
Blood. Serum or plasma measurement of Aβ has become possible and reliable thanks to the increased sensitivity and specificity of the available diagnostic platforms. Initial testing was based on ELISA (enzyme-linked immunosorbent assay), but results have been equivocal at best [148,168]. Interference by plasma proteins was likely significant in these early attempts. ELISA have been progressively improved to reach higher sensitivity, and other platforms have been developed, including SiMoA (Single Molecule Array) [169,170,171], immunomagnetic reduction [172], multiplex assays (e.g., xMAP- [173]), MSD [174] and more. As discussed in more detail below, in order to overcome the complexity of the blood and the interference by blood proteins and heterophilic antibodies and low concentrations of the target biomarker [113], the more modern platforms work with very high dilution of the sample and amplification of the specific signal, allowing for detection of picomolar/mL (or less) concentrations of the target protein [175,176]. Thanks to these advances, we can now reliably measure the Aβ42/40 ratio in plasma.
Studies using SiMoA have, first of all, confirmed a significant correlation between CSF and plasma [170,177] Aβ42, Aβ40 and their ratio, and further confirmed the CSF data on AD and MCI vs. control patients; AD and MCI patients show reduced levels of Aβ42 and Aβ42/40 ratio compared to cognitively intact control subjects, confirmed by amyloid PET.
High-precision assays for plasma Aβ42/40, using immunoprecipitation followed by liquid chromatography-mass spectroscopy (IP–MS) [178], are strongly predictive of brain amyloidosis as validated by benchmark amyloid-β-PET, and reach 90% accuracy [175,179,180]. In one study, a novel fragment ratio to Aβ42 (APP669–711/Aβ42) was also used to predict amyloid-β-PET positivity [175].
Interestingly, in the case of Aβ, another factor that interferes with the accurate plasma measurement is its extra-cerebral production (e.g., platelets, [181]), which is difficult to control. Hence, while it is possible to detect a reduction of the Aβ42/40 ratio in plasma in AD vs. control subjects, plasma reduction only totals about 15%, while in CSF we can see as much as 50% reduction [115,176]. When analyzing the overall trajectory of Aβ variation in plasma vs. CSF, in parallel with Aβ deposition in the brain by PET, by a variety of modern assays, plasma Aβ does not perform as well as CSF [182], showing a narrower range, making it still a less sensitive tool.

2.2. Tau

In AD, tau undergoes significant conformational change and abnormal phosphorylation, leading to the formation of inclusions throughout the brain, that start in the entorhinal cortex (EC) and, as the disease evolves, spread progressively throughout the brain, leading to synapse loss and neuronal death, which correlate well with the clinical onset and progression of symptoms [25,183,184]. High levels of tau, both total (t-tau) and phosphorylated (p-tau), have consistently been detected in CSF of AD patients compared to healthy controls [115,185,186,187,188,189,190,191,192]. Studies in FAD and SAD suggest that the tau-PET signal associated with neurofibrillary tangle pathology increases at symptom onset and appears about one decade after CSF soluble tau increase [193,194,195,196]. Other studies suggest that, while neurodegeneration progresses, the rate of increase of p-tau and t-tau levels may actually be reduced [118,133,197], likely in relation to the massive neuronal loss. In this dynamic scenario, where the rate of p-tau accumulation changes, and Aβ may show fluctuations in time, as discussed previously, an ideal biomarker should be specific enough for an adequate pre-clinical detection of AD and to track the disease progression.
As previously reported by Augustinack et al. [183], distinct tau phosphorylation sites correlate with severity of neuronal cytopathology in AD. Pre-tangle state, intra- and extra- neuronal neurofibrillary tangles were used to characterize different stages of pathology. More specifically, p-T231, p-S262, and p-T153 (T: Threonine, S: Serine) were shown to stain non fibrillar tau and were identified as early phosphorylation markers; p-T175/181, p-S262/p-S356, p-S422, p-S46 and p-S214 co-localized with intra-neuronal fibrillar tau structures, and increase as disease progresses; p-S199/p-S202/p-T205, p-T212/p-S214 and p-S396/p-S404 identified instead extracellular tangles composed of substantial filamentous tau structures, seen in later stages of the disease.
In addition to focusing on crucial site-specific phosphorylation, it has been shown, by cryo-EM analysis in AD as well as in other tauopathies, that the most disease-specific change is a tridimensional folding of the tau filaments, which is a signature of the particular disease examined; these tau molecular conformers can in fact differentiate between AD, Pick’s disease and corticobasal degeneration (CBD) [198,199,200,201]. Moreover, an AD-specific epitope formed by two discontinuous portions of tau, 7EFE9 and 313VDLSKVTSKC322 and recognized by the MC1 antibody [198,202,203], represents an early aberrant conformation of tau present both in a soluble form of the protein and in paired helical filaments (PHF) assemblies [204]. Interestingly, the level of MC1 reactivity correlates with the severity and progression of AD. To our knowledge, no assay has been developed to detect this early marker of pathology in biofluids, likely due to the possibility that tau truncated species [205,206,207,208] could prevent the identification of a conformational tau epitope using the currently available immunoassays, which are-based primarily on mid-region-directed antibodies. We believe that, although this pathological feature is not per se easily “translatable”, it should be kept in consideration when designing and developing new tau biomarkers platforms.
In summary, the identification of precise phosphorylated tau species and conformers in the biological fluids will contribute not only to early preclinical diagnosis of AD, but will also allow for a “personalized” approach by staging the disease in its progression and possibly monitoring the effect of therapeutic interventions. Here below is a review of the classic and novel approaches to detect tau and its phosphorylated modifications in CSF and blood.
CSF. Increased phosphorylation and axonal degeneration are believed to induce passive release of tau from the microtubules into the extracellular space, possibly as a neuronal response to Aβ toxicity [209,210]. This process results in augmented levels of t-tau and p-tau in the interstitial fluid and consequently in the CSF. From a neuropathological perspective, analysis of postmortem brains has shown a certain degree of correlation between the neocortical tau burden and CSF p-tau [190,211]. Tau phosphorylated at Threonine 181 (p-T181), t-tau and Aβ42 have been extensively validated in the CSF as biomarkers of AD and are currently widely used as diagnostic benchmarks in clinical and research studies [54,109,212]. CSF p-T181 tau has also proven useful in differential diagnosis of dementia [213,214,215] and to predict cognitive decline in preclinical and prodromal disease stages [213,216,217], with specificity for AD.
An ongoing effort focuses on standardizing protocols and developing criteria for the appropriate use of CSF tau assessment in the diagnosis of AD, with the intent of integrating CSF biomarkers into clinical practice [218,219,220]. In this respect, the distinctive value of the CSF p-T181 tau level has still to be thoroughly demonstrated. In one comparison study, p-T181, tau phosphorylated at Serine 199 (p-S199), and tau phosphorylated at Threonine 231 (p-T231) showed similar diagnostic accuracies for AD [221]. On the other hand, Spiegel et al. [222] reported that CSF p-T231 tau levels are superior to p-T181 in clustering AD subjects from normal control, suggesting the potential for CSF p-T231 assay to be employed in early AD diagnosis. These data are in line with the AD neuropathological picture where p-tau231 shows greater sensitivity for neurofibrillary tangles than p-T181 [211,223], with propensity to accumulate in layer II of the entorhinal cortex [183], known to be the first limbic area in AD consistently targeted by neurofibrillary pathology [224].
Together with p-T181 and p-T231, other phospho-epitopes are under investigation with the hope to find even better AD diagnostics. Barthélemy et al. [225] reported that CSF tau phosphorylated at Threonine 217 (p-T217) is specifically associated with amyloid-β pathology, suggesting a strong interplay between AD amyloidosis and hyperphosphorylation of tau on p-T217; moreover, p-T217 outperforms p-T181 as a biomarker for detecting both the preclinical and advanced forms of Alzheimer’s disease. In a second study, Barthelemy et al. [226] suggested a pattern of tau staging linking CSF site-specific phosphorylation to three different phases of the disease progression. By analyzing FAD, SAD and patients with unaffected cognition but at risk of disease (based on the presence of abnormal Aβ pathology); the authors identified an array of tau phosphorylation uniquely associated with structural, metabolic, neurodegenerative and clinical markers of disease. The study shows that (1) p-T217 and p-T181 rise in parallel to aggregated Aβ as early as 20 years before the development of neurofibrillary tangles, (2) p-T205 correlates with hypometabolism and atrophy closer to symptom onset, and (3) t-tau spikes when cognitive decline manifests. All of these data are in line with another study where the associations of p-T217 with tau PET, CSF and PET measures of amyloid pathology were tested in comparison to p-T181 [220]; the authors found that CSF p-T217 correlates better than p-T181 with PET measures of tau and amyloid pathologies in AD, and more accurately distinguishes AD from non-AD neurodegenerative disorders, suggesting the benefit of employing CSF p-T217 as a biomarker of AD in clinical practice.
Furthermore, investigating tau fragments in CSF, Chen et al. [227] developed a set of tau immunoassays able to detect different populations of tau fragments in CSF and blood. The authors were able to demonstrate that mid-region- and N-terminal-containing fragments increase with disease, while full-length tau is just a small fraction of the tau present in both normal and AD CSF. A recent study has also shown that the ratio between C-terminally truncated tau368 and t-tau is significantly decreased in patients with Alzheimer’s disease, with a strong correlation with 18F-GTP1 retention in brain [207] (a measure of tangle pathology in vivo); these data suggest that tau fragment may preferentially accumulate in tangles, and the CSF ratio tau368/t-tau reflects tangle pathology in brain and can be used as an additional tau biomarker to stage and improve the diagnosis of Alzheimer’s disease.
Several studies indicate that TBI can be classified as an independent risk factor for developing Alzheimer’s disease. As previously discussed for Aβ, the axonal injury resulting from the trauma drives the development of both Aβ plaques and NFT [228,229,230,231,232]. Interestingly, not only do tau levels increase in the CSF of the concussed patients undergoing repetitive trauma (i.e., sports players), but serum t-tau levels have been reported to increase in the acute stage following the concussion, while decreasing during rehabilitation [230,233,234]. Similarly, serum p-tau (p-T231 and p-S202) levels, though reduced, were shown to be still higher compared to the control levels even at six months post-injury [235].
When considering tauopathies different from AD with significant NFT pathology and neurodegeneration, CSF p-T181 and t-tau levels do not necessarily increase; this is the case for progressive supranuclear palsy (PSP) and some forms of FTD [221,236,237,238]. Why increased CSF t-tau and p-tau are specific to AD can be explained by considering that (1) the pattern of tau phosphorylation occurring in these disorders is disease-specific, (2) tangle pathology and neurodegeneration in AD may be consequent to Aβ accumulation, with disease-specific molecular cascades, such as kinases and proteases activation, (3) disease-specific tau truncation could prevent detection by currently available assays, and (4) specific pathological changes can be more severe in AD than in other tauopathies.
Blood. Until recently, AD biomarkers’ quantification was limited to the CSF. However, while combining CSF biomarker analyses and PET scans have the potential to improve diagnostic accuracy, these methodologies show limitations, impeding their use as first-line diagnostics [113]. Developing an accurate and standardized blood test will be more cost effective than PET imaging [239,240], less invasive than CSF testing [241] and faster. Hence, blood-based biomarkers represent next-generation diagnostics for AD and other CNS diseases. One matter of concern is that blood tau has the potential to derive from other sources in addition to neuronal tau [242,243]. In this respect, novel ultrasensitive detection technologies have been developed to facilitate the detection of very low concentrations and variations of tau in the blood [186,244,245] using very high dilutions of the biological fluid: employing these technologies has pushed forward the potential for plasma tau to serve as a biomarker for neurodegeneration in AD, avoiding the interference of non-neuronal tau.
Longitudinal studies originally showed that higher levels of plasma t-tau are associated with greater cognitive decline and risk of MCI [246,247,248], with no correlation with elevated brain Aβ [247]. As discussed, plasma t-tau assays can identify neuronal injury in acute brain disorders, such as TBI, but work relatively poorly for AD diagnosis, and the correlation with CSF t-tau is weak [249]. In this context, t-tau could be intended as a non-specific prognostic marker for dementia and related endophenotypes [248]. Despite the lack of correlation between plasma and CSF t-tau, Fossati et al. [186] have recently demonstrated that by combining plasma t-tau with CSF t-tau or CSF p-T181 tau, the accuracy for the differentiation of AD versus cognitively normal controls increases, suggesting that plasma tau may be a useful biomarker to increase diagnostic power and determine therapeutic efficacy in clinical studies.
Analysis of phosphorylated tau in blood has so far been focused on the development and validation of assays for p-T181 [182,246,250,251,252,253], demonstrating that increased plasma p-T181 (1) correlates with amyloid and tau PET measures [250,254,255], allowing the identification of individuals with Aβ pathology but tau PET-negative measurements [253], (2) goes in parallel with CSF p-T181 [182,255], and (3) can help in the differential diagnosis of non-AD types of dementias [253,254,255].
While writing this review, two groups published strong data in support of the use of a blood-based assay detecting p-T217 tau. First, Barthelemy et al. [256] developed a protocol of purification and enrichment of tau from plasma, followed by liquid chromatography (LC-MS); this system (1) allows reliable detection of p-T217 showing high correlation with the CSF measurements, and (2) confirms the ability of this biomarker in clustering amyloid positive and negative patients. The limitation of this strategy is the high volume of blood needed to purify and extract tau and the elaborate methodology, not compatible with the idea of developing a fast and easy-to- use assay. In the second study, Palmqvist et al. [256] employed the Meso Scale Discovery (MSD)/Lilly platform (described below) and validated the value of p-T217 in plasma to discriminate Aβ+ MCI and AD patients from other neurodegenerative disorders and controls, including sensitive discrimination of autosomal-dominant pre-symptomatic Aβ+ patients. In all, given the high potential of measuring CSF p-T217 previously described, it will be of great interest to follow the development and validation of novel ultrasensitive platforms aimed at detecting p-T217 in blood, employing fully automated platforms to integrate this biomarker in clinical practice. Similarly, setting up sensitive and specific assays able to detect p-T231 (early phosphorylated epitope) and p-S396/404 (late phosphorylated epitope) in blood will also be relevant to stage the AD pathology and to monitor future therapeutic interventions.
Finally, an ultra-sensitive assay was developed in blood, to detect an N-terminal tau fragment (NT1) [227], showing that N-terminal fragments of tau mapping in the 6–198 sequence are increased in AD and MCI patients and may be useful as a screening blood-based test for AD. This approach is extremely significant due to the possible generation of disease-specific tau fragments (as for APP and Aβ), and will need further investigation.
Immunoassays and ultrasensitive platforms to detect tau in biological fluids. Several assays have been tested and extensively validated in CSF and plasma. In Table 1 we select and catalogue the most used assays in the tau biomarker field at the moment, with emphasis on their specificity, fluid samples’ applicability and limit of detection. The list of assays includes (1) standard and second generation ELISAs (INNOTEST, EUROIMMUN, Elecsys), (2) MSD platforms (3) Single-Molecule enzyme-linked immunosorbent assay (SiMoA), (4) Mass spectrometry analysis, (5) superconducting-quantum-interference-device Immunomagnetic reduction (SQUID-IMR). The detection range spans from few pg/mL to fg/mL according to the sensitivity of the assay. EUROIMMUN and Elecsys are automated second-generation immune assays that have demonstrated good analytical performance with clinically relevant measuring ranges, supporting their use in clinical trials and practice [257,258]. MSD technology with electrochemiluminescence detection comes with several advantages compared to standard immunoassays, including high sensitivity, low background and wide dynamic range of detection [174]. Notably, Palmqvist et al. [182] recently ran a comparative analysis of Aβ and tau CSF assays from five different manufacturers: Elecsys (Aβ42, Aβ42/40, t-tau, p-tau181) EUROIMMUN (Aβ42, Aβ42/40, t-tau, p-tau181), INNOTEST (Aβ42, Aβ42/40, t-tau, p-tau181), MSD (Aβ42, Aβ42/40, t-tau) and Lilly (p-tau181, p-tau217): Aβ42, Aβ42/40, p-tau and t-tau had very similar trajectories between assays.
Moving to ultra-sensitive platforms, Rissn et al. previously developed [169,259] a Single-Molecule enzyme-linked immunosorbent assay (SiMoA) that detects serum proteins at sub-femtomolar concentrations, restricting the fluorophores produced by individual enzymes to considerably small volumes, hence capturing the fluorescence of each single molecule. This methodology has been extensively used to improve t-tau and p-tau detection in CSF and plasma (Quanterix).
SiMoA and INNOTEST ELISA have been also demonstrated to strongly correlate in CSF t-tau in a comparison study [186]. Moreover, in a recent study, Karikari et al. [253] validated a blood-based immunoassay measuring p-T181 using a sandwich immunoassay format on SiMoA technology.
To further quantify tau at high sensitivity, Barthelemy et al. has developed a protocol to detect tau and p-tau in CSF and plasma employing targeted high-resolution mass spectrometry (MS) [225,226,256,260,261]. Additionally, superconducting-quantum-interference-device Immunomagnetic reduction (SQUID-IMR) has proven efficient at quantifying t-tau and p-T181 in plasma in early stages of AD [172,252].

3. Biomarkers of Synaptic and Neuronal Loss

Synaptic loss first and overt neuronal demise later are fundamental aspect of AD pathology and are, together with tau deposition, the real correlate of the manifestation of symptoms. Neurodegeneration is the “N” in the ATN system, and is a fundamental correlate of the functional disability of AD patients. When patients present to their doctors with memory loss or other cognitive changes, due to AD, it is estimated that a large number of neurons have already been lost to neurodegeneration, but recent models are looking at the effect of more subtle synaptic loss as an inciting event responsible for symptoms [70]. In this context, several markers of synaptic disruption have been proposed as indicators of disease onset and progression.

3.1. Neurofilament Light Chain (NfL)

NfL belongs to the type IV intermediate filaments, which make up the neuronal axoskeleton, maintain neuronal caliber and play a role in intracellular transport to axons and dendrites. With the neurodegenerative process, in AD as in other disease [262], NfL is released in the extracellular space. NfL has been detected in both CSF and blood, and appears to be extremely sensitive to AD onset and progression, predicting Aβ PET positivity as well; furthermore, longitudinal studies in familial AD, have shown accurate prediction of disease onset, as far as 10 years prior to symptoms [263,264,265,266]. The tight correlation between CSF and blood values [267] make NfL one of the most reliable blood biomarkers to predict onset and course of neurodegeneration.

3.2. Neurogranin

Neurogranin (Ng) is a neural-specific postsynaptic protein, concentrated on dendritic spines, in the hippocampus and basal forebrain. Given its localization at the epicenter of the pathogenic events in AD, it is not surprising that it would be released in the extracellular space and be detected in CSF. In AD, CSF levels of Ng grow quite rapidly prior to the presentation of cognitive deficits, before NfL does, in an Aβ-dependent way [268]. Plasma levels do not seem to be as reliable as CSF, possibly due to peripheral production [269]. Importantly, and contrarily to NfL, other neurodegenerative disease do not show an increase of Ng, raising the possibility of an AD-specific synaptic effect [185].
Other biomarkers in this category are being investigated, such as NSE and SNAP-25, and are the subject of extensive analysis, but are mostly used in complex and integrated predictive models [133,270] and will not be reviewed here.

4. Biomarkers of Inflammation and Microglia “Dysfunction”

Inflammation, microglia and astrocytes activation are well-recognized features of AD pathology, as for other neurodegenerative diseases. It is unclear if they are integral part of the original pathogenesis or are reactive features to the deposition of amyloid and tau. To really understand this aspect, we should investigate, with an unbiased approach, the very early features of patients with a predisposition to develop AD pathology, for example in large autosomal dominant cohorts, in prospective longitudinal studies, and some effort is under way in this context. For the time being, we will focus on the data available from the earliest detectable stages of AD pathology, the prodromal AD cases.

4.1. Soluble TREM2

The Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) is a transmembrane receptor, belonging to the family of cell surface transmembrane glycoproteins [271] which mediates signaling and cell activation following ligand binding. It is expressed and functions in subsets of myeloid cells, e.g., granulocytes, dendritic cells, macrophages, and microglia in the brain [272,273,274,275]. Among its complex and protean roles, its function is to regulate immune cells’ activation upon binding of a variety of ligands [66], including APOE and Aβ. In the brain, where it may be expressed preferentially in some regions, such as the hippocampus, and differentially in response to the pro/anti-inflammatory environment, it mediates microglia activation, proliferation, migration, apoptosis and expression of pro-inflammatory cytokines, in particular after binding with aggregated proteins such as Aβ, and facilitates the phagocytosis of apoptotic neurons [276]. TREM2 expression is increased in many neurodegenerative conditions, stroke, trauma and even ageing. Rare variants of TREM2, discovered by GWAS studies [277,278], have been associated with the risk of developing AD, in the range of the risk conferred by APOE4, of 2- to 4-fold (heterozygous). The most common AD-associated variant recognized in populations of Europeans descent, but not in Chinese and African-Americans, is the R47H [277,279]. This TREM2 mutant is associated with earlier onset of disease and higher CSF tau levels [280] and seems to demonstrate reduced activity in ligands-binding and hence signal transduction, with a net result of reduced microglia activation and, in AD models, less clearance of Aβ [274]. Given this premise, it is only natural to look at TREM2 as a possible biomarker for AD. The soluble form of TREM2 (sTREM2) is found in both CSF and blood [281,282]. In CSF, sTREM2 is elevated in AD patients vs. controls, with positive association with t-tau levels [212,283]. In familial AD, sTREM2 CSF levels increase prior to clinical onset, but after detectable changes of Aβ and tau in CSF [284]; this is a moment in time where the brain is already at an advanced stage of pathology, in particular tau, with significant neuronal loss and when patients are soon going to lose their cognitive reserve and show symptoms. From a clinical perspective, this is relatively late to be useful as early prediction of disease or point of intervention, but from a pathogenic point of view, it may signify a “breaking point” of the Aβ and tau pathology containment system, where microglia cannot keep up with plaques and tangles any more. This is extremely important to consider in the development of therapeutic strategies in the future. More work is needed to create more sensitive assays in order to detect sTREM2 blood changes, as early as possible. Additionally, since the degree of inflammation and microglia activation differs from patient to patient, and secondary to treatment (e.g., anti-amyloid strategies), a sensitive blood sTREM2 assay may be a very useful tool to monitor phenotypic variants and adverse events form upcoming treatments.

4.2. YKL-40

YKL-40 is a glycoprotein expressed in both astrocytes and microglia, and is the protein product of the CHI3L1 gene, a.k.a., Chitinase 3 Like 1. YKL-40 actually lacks chitinase activity and is secreted by activated macrophages, chondrocytes, neutrophils and synovial cells, and plays a role in inflammation and tissue remodeling [285,286,287,288]. The first indication that YKL-40 may be relevant for AD came from an aprioristic evaluation of potential novel AD biomarkers [289]. Since then, several studies have now looked at the both CSF and plasma levels of this protein. YKL-40 is elevated in AD vs. control subjects [106,289,290], although it was also significantly elevated in patients with FTD [291]. Similarly, in MCI, baseline higher levels of YKL-40 were associated with significant more risk to progress to AD [106,289] with a tendency to continue to raise with disease progression [182]. The CSF ratio of YKL-40/Aβ42 was shown to be predictive of the onset of cognitive symptoms [289]. Plasma levels of YKL-40 have shown similar trends, but further validation may be needed to confirm reliable use [292]. In all, YKL-40, with its trend toward increase with disease progression, can be an associated marker for progression and prognosis.

4.3. Other Biomarkers of Inflammation and Glial Activation

Several pro- and anti-inflammatory factors have been looked at in both CSF and blood, including IL1, IL6, TNFα, VILIP-1, sTNFR and IP-10 [293,294,295], with some correlation to AD and MCI. Their significant is still unclear, and some are now part of multiplex panes, used in association with specific AD markers, arguably as prognostic indicators of disease progression.
GFAP (glial fibrillary acidic protein) is one of the major intermediate filament proteins of mature astrocytes, and it is released by astrocytes during neurodegeneration [296]. It is not properly a marker of inflammation, but it is a sign of a reactive brain milieu, as astrocytes tend to proliferate in response to tissue injury, and eventually release large amounts of GFAP in the interstitial fluid. Recently, the astrocytic response to brain damage and the inflammatory microglia has been characterized [297,298,299], and is very relevant in the context of AD pathology. The currently available CSF GFAP data show a correlation between increased levels and Aβ and tau evidence of AD [115,300], and recent data on plasma GFAP showed increased concentrations in late and early onset AD patients compared with cognitively normal controls [301]. Given the non-specific nature of GFAP, its use may be in the future confined to monitoring disease onset and progression, and possible response to treatment.
In all, given both the convergent and divergent nature of the inflammatory response, which involves a myriad of molecules and pathways, leading to a fine balance between protective and deleterious inflammation, and the fact that inflammation is an unavoidable element of ageing, any single marker would not be useful in isolation. Rather, a comprehensive approach using combinations of such factors, associated with specific marker of AD and markers of synaptic/neuronal loss (see below), has the best chance to be useful in clinical practice and research.

5. Biomarkers of Other Associated Pathology

5.1. TAR DNA-Binding Protein 43 (TDP-43)

TDP-43 can be present as co-pathology in AD (up to 50% of cases), and is the main feature of other neurodegenerative diseases, such as FTD and ALS. Its role is that of a transcriptional repressor, which regulates genes’ alternate splicing. An extensive study [302] was able to link the presence of TDP-43 in AD brains with the likelihood of clinical AD presentation; but to date, a reliable CSF or blood marker is not available, although notable attempts and data are present for ALS [303,304]. Part of the issue is the non-specificity of TDP-43 accumulation, as the concept of a spectrum of TDP-43 neurodegenerative presentation is becoming more evident [305]. In this context, TDP-43 may be useful as a biomarker in the future to predict course of disease or variants of AD presentation.

5.2. Alpha-Synuclein

Alpha-synuclein is the key component of Lewy bodies, and is classically found in Parkinson’s pathology, LBD, multi-system atrophy (MSA) and AD (also 50% of cases) variants. It is involved in presynaptic signaling, inhibiting phospholipases and moderating vesicles transport at the terminals [306]. As for TDP-43, despite the feasible measurement of α-synuclein in CSF and blood, no significant correlation has been made to date, likely due to the non-specific nature. What is different here is the possibility to harness α-synuclein’s specific tendency to aggregate in a “prion-like” fashion [307,308], for example by CSF RT-QuIC (real-time quaking-induced conversion), likely in virtue of its conformational changes [309]. Further studies will have to determine the sensitivity of the test for AD, as being able to detect synucleinopathy in AD cases would be highly significant for prognostic, therapeutic and research purposes.

6. Vascular Damage and the Blood Brain Barrier

Vascular damage is an increasingly recognized pathological component of AD, both as an independent consequence of the toxicity of Aβ and tau [310,311], and as a frequent co-morbidity [312,313] with microvascular disease. The diagnosis if “mixed dementia” is ever more frequent, as we scan more patients in our clinical practice. Although the real significance of the interplay between amyloid/tau pathology and the microvascular damage is still unclear, as the basic pathological mechanisms are quite complex, we have been able to detect vascular damage and blood-brain barrier (BBB) leak [314,315] in vivo. Modern magnetic resonance imaging (MRI) techniques, such as dynamic contrast-enhanced (DCE)-MRI, can show BBB permeability in specific regions, i.e., the hippocampus [315,316]. This abnormal permeability seems to be proportional to ageing [317], and is more frequent in MCI and AD patients compared to unimpaired control subjects [315,318,319]. An interesting correlate of MRI studies is the use of a marker of brain capillary damage, the soluble platelet-derived growth factor receptor-β (sPDGFRβ), which is highly expressed in brain capillary pericytes and vascular smooth muscle cells [315], and is shed in the CSF after brain injury, possibly in relation of the activation of proteases such as ADAM10 [320]. Emerging data show that ageing and cognitive decline, independently of AD pathology, are associated with increased CSF levels of sPDGFRβ, which correlate with MRI permeability [317]. Although the use of BBB permeability diagnostics is still in its infancy, and the specificity for AD pathology is unclear, it would certainly be a precious prognostic tool in the hands of clinicians. In this context, and somehow beyond the scope of the current manuscript, the use of gadolinium-free MRI techniques to assess BBB permeability (K-trans) would be extremely welcome.

7. Multi-Target Platforms

With so many biomarkers coming onto the scene, and in order to advance both the diagnostic and prognostic assessments, a new generation of assays is under development with the goal to simultaneously catch multiple targets within the same fluid sample. Using such types of assays will be extremely informative in order to identify AD co-pathologies, characterize disease progression and monitor treatment response. Table 2 contains a list of the more popular, commercially available, multi-target platforms.
The NeuroToolKit, in development with Roche’s Elecsys electroluminescence immunoassay platform [300], includes (1) α-synuclein as a marker of synaptic dysfunction, (2) S100b, YKL-40, and glial fibrillary acidic protein (GFAP) as markers of astrocyte activation, (3) sTREM2 and IL-6 as markers of microglial activation and inflammation, and (4) NfL and neurogranin as markers of axonal injury and synaptic dysfunction.
Simultaneous measurement of Aβ42, t-tau and p-T181 in CSF has been previously developed by the xMAP technology [321]. The MILLIPLEX® map multiplex immunoassay kit is a 4Xplex panel enables the simultaneous measurement of Aβ1-40, Aβ1-42, t-tau and p-T181 in human CSF.
The NEUROLOGY 3-PLEX A (SiMoA, Quanterix) detects t-tau, Aβ1-40 and Aβ1-42 in both CSF and blood.
The NEUROLOGY 4-PLEX A (SiMoA, Quanterix) detects t-tau, NfL, GFAP and ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1) both in CSF and blood.
A new 4-PLEX assay has been recently developed (SiMoA, Quanterix) with the ability to target Aβ1-40, Aβ1-4, GFAP and NfL in both CSF and blood (Alzheimer Association International Conference, 2020. Available online: https://alz.confex.com/alz/20amsterdam/meetingapp.cgi/Paper/43506. Accessed on 29 July 2020), and was validated against the parent assays. We await studies on larger number of patients, with diagnostic and prognostic implications.
Using a single platform with the ability to test multiple targets, such as in this case, would be invaluable in clinical practice, given how precious CSF and even blood samples are. Ideal kits should include markers specific of disease (e.g., Aβ42/40 ratio, p-T217 tau) and biomarkers of severity of disease, as NfL, sTREM2 etc.

8. Immunoassays, Ultrasensitive Platforms and Their Potential Limitations

One main concern in the development of an immunological assay is to exclude any factor that could potentially (1) interfere with the detection of the analyte and (2) alter the antibody binding. The first category includes pre-analytical factors (sample storage, anti-coagulants), anti-analyte antibodies, and hormone binding proteins that can change the measurable concentration of the analyte in the sample; the second group consists instead of antibodies such as heterophile antibodies, human anti-animal antibodies (HAAA), and human anti-mouse antibodies (HAMA) which are found in the biological fluids and are able to interfere with the antibody binding in the assay [322]. Unlike plasma (~7 g protein/100 mL) the CSF has only ~0.025 g protein/100 mL—mainly albumin [323]. Given the different composition of CSF and blood, it is relatively easy to anticipate the different susceptibility to assay interference of these two different biological fluids. While the CSF is a relatively “clean” fluid with respect to protein concentration, plasma contains heterophilic antibodies and other molecules that might interfere with the measurements of a biomarker of interest [245]. The presence of HAMA in human plasma has been described in 30% of the population, and consequent interference by human anti-globulin antibodies in immunoassay has been reported in several studies [324,325,326,327,328]. For these reasons, detecting biomarkers for CNS disease in blood not only calls for highly sensitive and specific assays, but require to efficiently remove all the potential confounders. Different methods for the reduction of heterophile antibody and anti-animal interferences in immunoassays have been previously described and include ways to remove or block the interfering antibody [322,329,330]. In this respect, our group has previously developed a protocol combining NaOAc (sodium acetate) and heat-treatment to ‘purify’ tau from the blood of transgenic animals undergone tau immunotherapy [328]. This protocol was validated using mouse and human serum. While the NaOAc/heat-treatment did not affect tau reactivity on CSF samples, human plasma samples displayed an impressive reduction of the apparent tau signals after undergoing the heat extraction protocol. The use of the ultrasensitive assays described here has overcome some of these technical issues. In addition, the use of blockers against heterophilic antibodies in the sample diluent is also helpful to achieve reliable quantification [245]. We still believe that the previous considerations need to be considered, especially when analyzing human samples derived from patients undergone immunotherapy, where the risk of assay interference could compromise the interpretations of the disease course and therapy.
Ageing is per se associated with a reduction of the integrity of the BBB, due to a multitude of causes, e.g., prior infections, general inflammatory state, trauma, tumors and more [315,316]. MCI, AD and cognitive changes in ageing have been associated with non-specific BBB permeability in certain brain regions [317]. The resulting permeability may result in falsification of CSF data for reasons analogous to the blood.

9. Novel Approaches to Unbiased Biomarker Discovery: The Case for Metabolomics

A successful preclinical diagnosis of AD may benefit from an unbiased metabolomic analysis of biological fluids. Given that CSF metabolites reflect brain changes better than blood, CSF biomarkers are widely used in research and clinical practice [331]. The strength of metabolomics is in its ability to measure a plethora of metabolites in an unbiased manner, providing a snapshot of an individual’s biological status [331,332]. The ideal system would include a CSF metabolomic profile covering the whole lifespan of individuals belonging to both FAD and SAD cohorts, followed by plasma validation. While this analysis would require decades in human AD, or the longitudinal analysis of autosomal dominant families, running such studies on faithful animal models would make for an optimal surrogate, taking advantage of novel knock-in models, as opposed to transgenic ones, which reproduce the exact genetic defect found in humans [333,334,335,336,337,338].
Metabolomic studies have been performed in CSF, blood and post-mortem tissue from AD and MCI patients [331,339,340]. For example, elevated CSF glutamate and glutamine levels have been described in AD before [332,341,342], and other studies have shown blood and CSF metabolites changes in AD [343,344,345], some correlating with progression and risk of MCI to progress to AD [346]. Most identified pathways relate to energy metabolism, Krebs cycle, mitochondrial function, neurotransmitter and amino acids metabolism, and lipid biosynthesis. Longitudinal studies in FAD cases would be welcome in the near future to understand both the pathological relevance of the said pathways and their power to predict AD.

10. Discussion and Conclusions

Several studies have by now assessed the use of isolated biomarkers vs. their combinations to predict diagnosis of AD, or progression of MCI to AD. No single biomarker, in CSF, blood, imaging or cognitive alone is capable to predict onset and course reliably. Rather, the combination of multimodal biomarkers is required to reach reliable predictions [100,300,342,347,348]. CSF Aβ1-42 reduction (best proxied by the Aβ42/40 ratio) starts very early before symptoms onset, peaks a few years later when amyloid PET is at the zenith; CSF tau elevation follows Aβ, and represent clinical disease severity very well. NfL increase starts early, before tau, and is a very good indicator of neuronal damage early in the disease, though it is not specific for AD; still, its reliability in blood as well in CSF, makes it an excellent marker to track disease progression and possible response to therapies. Similarly, sTREM2 and YKL-40 are excellent markers of inflammation, and although their best use is in CSF, they could be extremely useful to monitor possible adverse events of therapeutic intervention. Aprioristic analysis of CSF and blood by metabolomic, proteomic, lipidomic and other large-scale approaches could be the key, if done early enough in the pathogenic course, to generate hypothesis on the primordial triggers of the disease, besides to identify AD patients and predict their trajectory. In this respect, autosomal dominant families and patients with Down’s syndrome [134,349,350] may be the ideal subjects for dedicated longitudinal studies.
Given this premise, the biomarker field has to drive every resources toward the development of blood-based assays able to provide high specificity and sensitivity to excel in both diagnosis (Aβ, and tau) and prognosis (i.e., NfL) of AD. Implementing AD biomarkers in clinical practice will require blood-based assays (1) easy-to-use in terms of human specimen collection (few mL of blood) and methodology, (2) with high sensitivity (under the pg/mL threshold) and specificity, (3) free of interference produced by several confounders associated with the ageing process. The focus on pre-symptomatic Aβ+ populations is paramount (e.g., autosomal-dominant cohorts) if we really want to diagnose the disease in a phase where significant intervention is still possible.

Author Contributions

Conceptualization, L.D., L.G., C.d.; methodology, C.d., L.G.; writing—original draft preparation, C.d., L.G.; writing—review and editing, L.D., L.G.; visualization, C.d.; supervision, L.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We would like to thank Peter Davies for his continued support and insight.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Apostolova, L.G. Alzheimer disease. Continuum Minneap. Minn. 2016, 22, 419–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hebert, L.E.; Weuve, J.; Scherr, P.A.; Evans, D.A. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology 2013, 80, 1778–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hebert, L.E.; Bienias, J.L.; Aggarwal, N.T.; Wilson, R.S.; Bennett, D.A.; Shah, R.C.; Evans, D.A. Change in risk of Alzheimer disease over time. Neurology 2010, 75, 786–791. [Google Scholar] [CrossRef] [PubMed]
  4. Fan, C.C.; Banks, S.J.; Thompson, W.K.; Chen, C.H.; McEvoy, L.K.; Tan, C.H.; Kukull, W.; Bennett, D.A.; Farrer, L.A.; Mayeux, R.; et al. Sex-dependent autosomal effects on clinical progression of alzheimer’s disease. Brain 2020, 143, 2272–2280. [Google Scholar] [CrossRef]
  5. Plassman, B.L.; Langa, K.M.; Fisher, G.G.; Heeringa, S.G.; Weir, D.R.; Ofstedal, M.B.; Burke, J.R.; Hurd, M.D.; Potter, G.G.; Rodgers, W.L.; et al. Prevalence of dementia in the united states: The aging, demographics, and memory study. Neuroepidemiology 2007, 29, 125–132. [Google Scholar] [CrossRef]
  6. Seshadri, S.; Wolf, P.A.; Beiser, A.; Au, R.; McNulty, K.; White, R.; D’Agostino, R.B. Lifetime risk of dementia and alzheimer’s disease. The impact of mortality on risk estimates in the framingham study. Neurology 1997, 49, 1498–1504. [Google Scholar] [CrossRef]
  7. Hebert, L.E.; Scherr, P.A.; McCann, J.J.; Beckett, L.A.; Evans, D.A. Is the risk of developing alzheimer’s disease greater for women than for men? Am. J. Epidemiol. 2001, 153, 132–136. [Google Scholar] [CrossRef]
  8. Chêne, G.; Beiser, A.; Au, R.; Preis, S.R.; Wolf, P.A.; Dufouil, C.; Seshadri, S. Gender and incidence of dementia in the framingham heart study from mid-adult life. Alzheimer’s Dement. 2015, 11, 310–320. [Google Scholar] [CrossRef] [Green Version]
  9. Carter, C.L.; Resnick, E.M.; Mallampalli, M.; Kalbarczyk, A. Sex and gender differences in alzheimer’s disease: Recommendations for future research. J. Womens Health 2012, 21, 1018–1023. [Google Scholar] [CrossRef]
  10. Altmann, A.; Tian, L.; Henderson, V.W.; Greicius, M.D.; Investigators, A.s.D.N.I. Sex modifies the apoe-related risk of developing alzheimer disease. Ann. Neurol. 2014, 75, 563–573. [Google Scholar] [CrossRef] [Green Version]
  11. Ungar, L.; Altmann, A.; Greicius, M.D. Apolipoprotein E, gender, and alzheimer’s disease: An overlooked, but potent and promising interaction. Brain Imaging Behav. 2014, 8, 262–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Yaffe, K.; Haan, M.; Byers, A.; Tangen, C.; Kuller, L. Estrogen use, apoe, and cognitive decline: Evidence of gene-environment interaction. Neurology 2000, 54, 1949–1954. [Google Scholar] [CrossRef] [PubMed]
  13. Kang, J.H.; Grodstein, F. Postmenopausal hormone therapy, timing of initiation, apoe and cognitive decline. Neurobiol. Aging 2012, 33, 1129–1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Barrett-Connor, E.; Laughlin, G.A. Endogenous and exogenous estrogen, cognitive function, and dementia in postmenopausal women: Evidence from epidemiologic studies and clinical trials. Semin. Reprod. Med. 2009, 27, 275–282. [Google Scholar] [CrossRef] [Green Version]
  15. O’Brien, J.; Jackson, J.W.; Grodstein, F.; Blacker, D.; Weuve, J. Postmenopausal hormone therapy is not associated with risk of all-cause dementia and alzheimer’s disease. Epidemiol. Rev. 2014, 36, 83–103. [Google Scholar] [CrossRef]
  16. Xu, J.; Murphy, S.L.; Kochanek, K.D.; Bastian, B.A. Deaths: Final data for 2013. Natl. Vital Stat. Rep. 2016, 64, 1–119. [Google Scholar]
  17. Murray, C.J.; Atkinson, C.; Bhalla, K.; Birbeck, G.; Burstein, R.; Chou, D.; Dellavalle, R.; Danaei, G.; Ezzati, M.; Fahimi, A.; et al. The state of us health, 1990–2010: Burden of diseases, injuries, and risk factors. JAMA 2013, 310, 591–608. [Google Scholar] [CrossRef] [Green Version]
  18. Gaugler, J.E.; Kane, R.L.; Kane, R.A. Family care for older adults with disabilities: Toward more targeted and interpretable research. Int. J. Aging Hum. Dev. 2002, 54, 205–231. [Google Scholar] [CrossRef]
  19. Schulz, R.; Czaja, S.J.; Lustig, A.; Zdaniuk, B.; Martire, L.M.; Perdomo, D. Improving the quality of life of caregivers of persons with spinal cord injury: A randomized controlled trial. Rehabil. Psychol. 2009, 54, 1. [Google Scholar] [CrossRef] [Green Version]
  20. Kasper, J.D.; Freedman, V.A.; Spillman, B.C.; Wolff, J.L. The disproportionate impact of dementia on family and unpaid caregiving to older adults. Health Aff. Millwood 2015, 34, 1642–1649. [Google Scholar] [CrossRef] [Green Version]
  21. Freedman, V.A.; Spillman, B.C. Disability and care needs among older americans. Milbank Q. 2014, 92, 509–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Anderson, L.A.; Edwards, V.J.; Pearson, W.S.; Talley, R.C.; McGuire, L.C.; Andresen, E.M. Adult caregivers in the united states: Characteristics and differences in well-being, by caregiver age and caregiving status. Prev. Chronic Dis. 2013, 10, E135. [Google Scholar] [CrossRef] [PubMed]
  23. Hurd, M.D.; Martorell, P.; Delavande, A.; Mullen, K.J.; Langa, K.M. Monetary costs of dementia in the united states. N. Engl. J. Med. 2013, 368, 1326–1334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Consensus recommendations for the postmortem diagnosis of alzheimer’s disease. The national institute on aging, and reagan institute working group on diagnostic criteria for the neuropathological assessment of alzheimer’s disease. Neurobiol. Aging 1997, 18, S1–S2.
  25. Braak, H.; Braak, E. Neuropathological stageing of alzheimer-related changes. Acta Neuropathol. 1991, 82, 239–259. [Google Scholar] [CrossRef] [PubMed]
  26. Hyman, B.T.; Phelps, C.H.; Beach, T.G.; Bigio, E.H.; Cairns, N.J.; Carrillo, M.C.; Dickson, D.W.; Duyckaerts, C.; Frosch, M.P.; Masliah, E.; et al. National institute on aging-alzheimer’s association guidelines for the neuropathologic assessment of alzheimer’s disease. Alzheimer’s Dement. 2012, 8, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Montine, T.J.; Phelps, C.H.; Beach, T.G.; Bigio, E.H.; Cairns, N.J.; Dickson, D.W.; Duyckaerts, C.; Frosch, M.P.; Masliah, E.; Mirra, S.S.; et al. National institute on aging-alzheimer’s association guidelines for the neuropathologic assessment of alzheimer’s disease: A practical approach. Acta Neuropathol. 2012, 123, 1–13. [Google Scholar] [CrossRef] [Green Version]
  28. Holtzman, D.M.; Mandelkow, E.; Selkoe, D.J. Alzheimer disease in 2020. Cold Spring Harb. Perspect. Med. 2012, 2, a011585. [Google Scholar] [CrossRef]
  29. Braak, H.; Braak, E. Evolution of the neuropathology of alzheimer’s disease. Acta Neurol. Scand. 1996, 165, 3–12. [Google Scholar] [CrossRef]
  30. Sengupta, U.; Nilson, A.N.; Kayed, R. The role of amyloid-β oligomers in toxicity, propagation, and immunotherapy. EBioMedicine 2016, 6, 42–49. [Google Scholar] [CrossRef] [Green Version]
  31. Goedert, M.; Spillantini, M.G. Ordered assembly of tau protein and neurodegeneration. Adv. Exp. Med. Biol. 2019, 1184, 3–21. [Google Scholar] [PubMed]
  32. Walsh, D.M.; Klyubin, I.; Fadeeva, J.V.; Rowan, M.J.; Selkoe, D.J. Amyloid-beta oligomers: Their production, toxicity and therapeutic inhibition. Biochem. Soc. Trans. 2002, 30, 552–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Goedert, M.; Eisenberg, D.S.; Crowther, R.A. Propagation of tau aggregates and neurodegeneration. Annu. Rev. Neurosci. 2017, 40, 189–210. [Google Scholar] [CrossRef] [PubMed]
  34. Ittner, A.; Ittner, L.M. Dendritic tau in alzheimer’s disease. Neuron 2018, 99, 13–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Stancu, I.C.; Vasconcelos, B.; Terwel, D.; Dewachter, I. Models of β-amyloid induced tau-pathology: The long and “folded” road to understand the mechanism. Mol. Neurodegener. 2014, 9, 51. [Google Scholar] [CrossRef] [Green Version]
  36. van der Kant, R.; Goldstein, L.S.B.; Ossenkoppele, R. Amyloid-β-independent regulators of tau pathology in alzheimer disease. Nat. Rev. Neurosci. 2020, 21, 21–35. [Google Scholar] [CrossRef]
  37. La Joie, R.; Visani, A.V.; Baker, S.L.; Brown, J.A.; Bourakova, V.; Cha, J.; Chaudhary, K.; Edwards, L.; Iaccarino, L.; Janabi, M.; et al. Prospective longitudinal atrophy in alzheimer’s disease correlates with the intensity and topography of baseline tau-pet. Sci. Transl. Med. 2020, 12, 5732–5745. [Google Scholar] [CrossRef]
  38. Franzmeier, N.; Neitzel, J.; Rubinski, A.; Smith, R.; Strandberg, O.; Ossenkoppele, R.; Hansson, O.; Ewers, M.; (ADNI), A.s.D.N.I. Functional brain architecture is associated with the rate of tau accumulation in alzheimer’s disease. Nat. Commun. 2020, 11, 347. [Google Scholar] [CrossRef] [Green Version]
  39. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [Green Version]
  40. Serrano-Pozo, A.; Mielke, M.L.; Gómez-Isla, T.; Betensky, R.A.; Growdon, J.H.; Frosch, M.P.; Hyman, B.T. Reactive glia not only associates with plaques but also parallels tangles in alzheimer’s disease. Am. J. Pathol. 2011, 179, 1373–1384. [Google Scholar] [CrossRef]
  41. Wake, H.; Moorhouse, A.J.; Jinno, S.; Kohsaka, S.; Nabekura, J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J. Neurosci. 2009, 29, 3974–3980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Vehmas, A.K.; Kawas, C.H.; Stewart, W.F.; Troncoso, J.C. Immune reactive cells in senile plaques and cognitive decline in alzheimer’s disease. Neurobiol. Aging 2003, 24, 321–331. [Google Scholar] [CrossRef] [Green Version]
  43. Serrano-Pozo, A.; Frosch, M.P.; Masliah, E.; Hyman, B.T. Neuropathological alterations in alzheimer disease. Cold Spring Harb. Perspect. Med. 2011, 1, a006189. [Google Scholar] [CrossRef] [PubMed]
  44. Perl, D.P. Neuropathology of alzheimer’s disease. Mt. Sinai J. Med. 2010, 77, 32–42. [Google Scholar] [CrossRef] [PubMed]
  45. Thal, D.R.; Schultz, C.; Botez, G.; Del Tredici, K.; Mrak, R.E.; Griffin, W.S.; Wiestler, O.D.; Braak, H.; Ghebremedhin, E. The impact of argyrophilic grain disease on the development of dementia and its relationship to concurrent alzheimer’s disease-related pathology. Neuropathol. Appl. Neurobiol. 2005, 31, 270–279. [Google Scholar] [CrossRef] [PubMed]
  46. Hansen, L.; Salmon, D.; Galasko, D.; Masliah, E.; Katzman, R.; DeTeresa, R.; Thal, L.; Pay, M.M.; Hofstetter, R.; Klauber, M. The lewy body variant of alzheimer’s disease: A clinical and pathologic entity. Neurology 1990, 40, 1. [Google Scholar] [CrossRef]
  47. Nelson, P.T.; Gal, Z.; Wang, W.X.; Niedowicz, D.M.; Artiushin, S.C.; Wycoff, S.; Wei, A.; Jicha, G.A.; Fardo, D.W. Tdp-43 proteinopathy in aging: Associations with risk-associated gene variants and with brain parenchymal thyroid hormone levels. Neurobiol. Dis. 2019, 125, 67–76. [Google Scholar] [CrossRef]
  48. Josephs, K.A.; Murray, M.E.; Whitwell, J.L.; Parisi, J.E.; Petrucelli, L.; Jack, C.R.; Petersen, R.C.; Dickson, D.W. Staging tdp-43 pathology in alzheimer’s disease. Acta Neuropathol. 2014, 127, 441–450. [Google Scholar] [CrossRef]
  49. Prasad, A.; Bharathi, V.; Sivalingam, V.; Girdhar, A.; Patel, B.K. Molecular mechanisms of tdp-43 misfolding and pathology in amyotrophic lateral sclerosis. Front. Mol. Neurosci. 2019, 12, 25. [Google Scholar] [CrossRef]
  50. Gómez-Isla, T.; Hollister, R.; West, H.; Mui, S.; Growdon, J.H.; Petersen, R.C.; Parisi, J.E.; Hyman, B.T. Neuronal loss correlates with but exceeds neurofibrillary tangles in alzheimer’s disease. Ann. Neurol. 1997, 41, 17–24. [Google Scholar] [CrossRef]
  51. Forner, S.; Baglietto-Vargas, D.; Martini, A.C.; Trujillo-Estrada, L.; LaFerla, F.M. Synaptic impairment in alzheimer’s disease: A dysregulated symphony. Trends Neurosci. 2017, 40, 347–357. [Google Scholar] [CrossRef] [PubMed]
  52. Overk, C.R.; Masliah, E. Pathogenesis of synaptic degeneration in alzheimer’s disease and lewy body disease. Biochem. Pharmacol. 2014, 88, 508–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Sabbagh, M.N.; Lue, L.F.; Fayard, D.; Shi, J. Increasing precision of clinical diagnosis of alzheimer’s disease using a combined algorithm incorporating clinical and novel biomarker data. Neurol. Ther. 2017, 6, 83–95. [Google Scholar] [CrossRef]
  54. Dubois, B.; Feldman, H.H.; Jacova, C.; Hampel, H.; Molinuevo, J.L.; Blennow, K.; DeKosky, S.T.; Gauthier, S.; Selkoe, D.; Bateman, R.; et al. Advancing research diagnostic criteria for alzheimer’s disease: The iwg-2 criteria. Lancet Neurol. 2014, 13, 614–629. [Google Scholar] [CrossRef]
  55. Boise, L.; Camicioli, R.; Morgan, D.L.; Rose, J.H.; Congleton, L. Diagnosing dementia: Perspectives of primary care physicians. Gerontologist 1999, 39, 457–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Beach, T.G.; Monsell, S.E.; Phillips, L.E.; Kukull, W. Accuracy of the clinical diagnosis of alzheimer disease at national institute on aging alzheimer disease centers, 2005–2010. J. Neuropathol. Exp. Neurol. 2012, 71, 266–273. [Google Scholar] [CrossRef]
  57. Cacace, R.; Sleegers, K.; Van Broeckhoven, C. Molecular genetics of early-onset alzheimer’s disease revisited. Alzheimer’s Dement. 2016, 12, 733–748. [Google Scholar] [CrossRef] [Green Version]
  58. Kimberly, W.T.; LaVoie, M.J.; Ostaszewski, B.L.; Ye, W.; Wolfe, M.S.; Selkoe, D.J. Gamma-secretase is a membrane protein complex comprised of presenilin, nicastrin, aph-1, and pen-2. Proc. Natl. Acad. Sci. USA 2003, 100, 6382–6387. [Google Scholar] [CrossRef] [Green Version]
  59. Van Broeckhoven, C. Presenilins and alzheimer disease. Nat. Genet. 1995, 11, 230–232. [Google Scholar] [CrossRef]
  60. Rademakers, R.; Cruts, M.; Van Broeckhoven, C. Genetics of early-onset alzheimer dementia. Sci. World J. 2003, 3, 497–519. [Google Scholar] [CrossRef]
  61. Sherrington, R.; Rogaev, E.I.; Liang, Y.; Rogaeva, E.A.; Levesque, G.; Ikeda, M.; Chi, H.; Lin, C.; Li, G.; Holman, K.; et al. Cloning of a gene bearing missense mutations in early-onset familial alzheimer’s disease. Nature 1995, 375, 754–760. [Google Scholar] [CrossRef]
  62. Levy-Lahad, E.; Wasco, W.; Poorkaj, P.; Romano, D.M.; Oshima, J.; Pettingell, W.H.; Yu, C.E.; Jondro, P.D.; Schmidt, S.D.; Wang, K. Candidate gene for the chromosome 1 familial alzheimer’s disease locus. Science 1995, 269, 973–977. [Google Scholar] [CrossRef] [PubMed]
  63. Welzel, A.T.; Maggio, J.E.; Shankar, G.M.; Walker, D.E.; Ostaszewski, B.L.; Li, S.; Klyubin, I.; Rowan, M.J.; Seubert, P.; Walsh, D.M.; et al. Secreted amyloid β-proteins in a cell culture model include n-terminally extended peptides that impair synaptic plasticity. Biochemistry 2014, 53, 3908–3921. [Google Scholar] [CrossRef] [PubMed]
  64. Riedel, B.C.; Thompson, P.M.; Brinton, R.D. Age, apoe and sex: Triad of risk of alzheimer’s disease. J. Steroid Biochem. Mol. Biol. 2016, 160, 134–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Yamazaki, Y.; Zhao, N.; Caulfield, T.R.; Liu, C.C.; Bu, G. Apolipoprotein e and alzheimer disease: Pathobiology and targeting strategies. Nat. Rev. Neurol. 2019, 15, 501–518. [Google Scholar] [CrossRef] [PubMed]
  66. Gratuze, M.; Leyns, C.E.G.; Holtzman, D.M. New insights into the role of trem2 in alzheimer’s disease. Mol. Neurodegener. 2018, 13, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Cai, Q.; Tammineni, P. Alterations in mitochondrial quality control in alzheimer’s disease. Front. Cell. Neurosci. 2016, 10, 24. [Google Scholar] [CrossRef] [Green Version]
  68. Devi, L.; Prabhu, B.M.; Galati, D.F.; Avadhani, N.G.; Anandatheerthavarada, H.K. Accumulation of amyloid precursor protein in the mitochondrial import channels of human alzheimer’s disease brain is associated with mitochondrial dysfunction. J. Neurosci. 2006, 26, 9057–9068. [Google Scholar] [CrossRef] [Green Version]
  69. Lamprecht, R.; LeDoux, J. Structural plasticity and memory. Nat. Rev. Neurosci. 2004, 5, 45–54. [Google Scholar] [CrossRef]
  70. Kashyap, G.; Bapat, D.; Das, D.; Gowaikar, R.; Amritkar, R.E.; Rangarajan, G.; Ravindranath, V.; Ambika, G. Synapse loss and progress of alzheimer’s disease—A network model. Sci. Rep. 2019, 9, 6555. [Google Scholar] [CrossRef] [Green Version]
  71. Fabiani, C.; Antollini, S.S. Alzheimer’s disease as a membrane disorder: Spatial cross-talk among beta-amyloid peptides, nicotinic acetylcholine receptors and lipid rafts. Front. Cell. Neurosci. 2019, 13, 309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Di Scala, C.; Yahi, N.; Boutemeur, S.; Flores, A.; Rodriguez, L.; Chahinian, H.; Fantini, J. Common molecular mechanism of amyloid pore formation by alzheimer’s β-amyloid peptide and α-synuclein. Sci. Rep. 2016, 6, 28781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Lasagna-Reeves, C.A.; Sengupta, U.; Castillo-Carranza, D.; Gerson, J.E.; Guerrero-Munoz, M.; Troncoso, J.C.; Jackson, G.R.; Kayed, R. The formation of tau pore-like structures is prevalent and cell specific: Possible implications for the disease phenotypes. Acta Neuropathol. Commun. 2014, 2, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Tseng, B.P.; Green, K.N.; Chan, J.L.; Blurton-Jones, M.; LaFerla, F.M. Abeta inhibits the proteasome and enhances amyloid and tau accumulation. Neurobiol. Aging 2008, 29, 1607–1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Myeku, N.; Clelland, C.L.; Emrani, S.; Kukushkin, N.V.; Yu, W.H.; Goldberg, A.L.; Duff, K.E. Tau-driven 26s proteasome impairment and cognitive dysfunction can be prevented early in disease by activating camp-pka signaling. Nat. Med. 2016, 22, 46–53. [Google Scholar] [CrossRef]
  76. Yamazaki, Y.; Kanekiyo, T. Blood-brain barrier dysfunction and the pathogenesis of alzheimer’s disease. Int. J. Mol. Sci. 2017, 18, 1965. [Google Scholar] [CrossRef] [Green Version]
  77. Venegas, C.; Kumar, S.; Franklin, B.S.; Dierkes, T.; Brinkschulte, R.; Tejera, D.; Vieira-Saecker, A.; Schwartz, S.; Santarelli, F.; Kummer, M.P.; et al. Microglia-derived asc specks cross-seed amyloid-β in alzheimer’s disease. Nature 2017, 552, 355–361. [Google Scholar] [CrossRef]
  78. Perea, J.R.; Llorens-Martín, M.; Ávila, J.; Bolós, M. The role of microglia in the spread of tau: Relevance for tauopathies. Front. Cell. Neurosci. 2018, 12, 172. [Google Scholar] [CrossRef] [Green Version]
  79. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [Green Version]
  80. Jack, C.R.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Feldman, H.H.; Frisoni, G.B.; Hampel, H.; Jagust, W.J.; Johnson, K.A.; Knopman, D.S.; et al. A/t/n: An unbiased descriptive classification scheme for alzheimer disease biomarkers. Neurology 2016, 87, 539–547. [Google Scholar] [CrossRef]
  81. Ekman, U.; Ferreira, D.; Westman, E. The a/t/n biomarker scheme and patterns of brain atrophy assessed in mild cognitive impairment. Sci. Rep. 2018, 8, 8431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ross, D.E.; Ochs, A.L.; Seabaugh, J.; Henshaw, T. Neuroquant® revealed hippocampal atrophy in a patient with traumatic brain injury. J. Neuropsychiatry Clin. Neurosci. 2012, 24, E33. [Google Scholar] [CrossRef] [PubMed]
  83. Brewer, J.B.; Magda, S.; Airriess, C.; Smith, M.E. Fully-automated quantification of regional brain volumes for improved detection of focal atrophy in alzheimer disease. AJNR Am. J. Neuroradiol. 2009, 30, 578–580. [Google Scholar] [CrossRef] [PubMed]
  84. Piccini, A.; Russo, C.; Gliozzi, A.; Relini, A.; Vitali, A.; Borghi, R.; Giliberto, L.; Armirotti, A.; D’Arrigo, C.; Bachi, A.; et al. Beta-amyloid is different in normal aging and in alzheimer disease. J. Biol. Chem. 2005, 280, 34186–34192. [Google Scholar] [CrossRef] [Green Version]
  85. Panza, F.; Lozupone, M.; Logroscino, G.; Imbimbo, B.P. A critical appraisal of amyloid-β-targeting therapies for alzheimer disease. Nat. Rev. Neurol. 2019, 15, 73–88. [Google Scholar] [CrossRef]
  86. Selkoe, D.J. Resolving controversies on the path to alzheimer’s therapeutics. Nat. Med. 2011, 17, 1060–1065. [Google Scholar] [CrossRef]
  87. Hane, F.T.; Robinson, M.; Lee, B.Y.; Bai, O.; Leonenko, Z.; Albert, M.S. Recent progress in alzheimer’s disease research, part 3: Diagnosis and treatment. J. Alzheimer’s Dis. 2017, 57, 645–665. [Google Scholar] [CrossRef] [Green Version]
  88. Orgogozo, J.M.; Gilman, S.; Dartigues, J.F.; Laurent, B.; Puel, M.; Kirby, L.C.; Jouanny, P.; Dubois, B.; Eisner, L.; Flitman, S.; et al. Subacute meningoencephalitis in a subset of patients with ad after abeta42 immunization. Neurology 2003, 61, 46–54. [Google Scholar] [CrossRef]
  89. Vellas, B.; Black, R.; Thal, L.J.; Fox, N.C.; Daniels, M.; McLennan, G.; Tompkins, C.; Leibman, C.; Pomfret, M.; Grundman, M.; et al. Long-term follow-up of patients immunized with an1792: Reduced functional decline in antibody responders. Curr. Alzheimer Res. 2009, 6, 144–151. [Google Scholar] [CrossRef] [Green Version]
  90. Lemere, C.A.; Masliah, E. Can alzheimer disease be prevented by amyloid-beta immunotherapy? Nat. Rev. Neurol. 2010, 6, 108–119. [Google Scholar] [CrossRef] [Green Version]
  91. Wiessner, C.; Wiederhold, K.H.; Tissot, A.C.; Frey, P.; Danner, S.; Jacobson, L.H.; Jennings, G.T.; Lüönd, R.; Ortmann, R.; Reichwald, J.; et al. The second-generation active aβ immunotherapy cad106 reduces amyloid accumulation in app transgenic mice while minimizing potential side effects. J. Neurosci. 2011, 31, 9323–9331. [Google Scholar] [CrossRef] [PubMed]
  92. Doody, R.S.; Thomas, R.G.; Farlow, M.; Iwatsubo, T.; Vellas, B.; Joffe, S.; Kieburtz, K.; Raman, R.; Sun, X.; Aisen, P.S.; et al. Phase 3 trials of solanezumab for mild-to-moderate alzheimer’s disease. N. Engl. J. Med. 2014, 370, 311–321. [Google Scholar] [CrossRef] [PubMed]
  93. Liu-Seifert, H.; Siemers, E.; Holdridge, K.C.; Andersen, S.W.; Lipkovich, I.; Carlson, C.; Sethuraman, G.; Hoog, S.; Hayduk, R.; Doody, R.; et al. Delayed-start analysis: Mild alzheimer’s disease patients in solanezumab trials, 3.5 years. Alzheimer’s Dement. N. Y. 2015, 1, 111–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Siemers, E.R.; Sundell, K.L.; Carlson, C.; Case, M.; Sethuraman, G.; Liu-Seifert, H.; Dowsett, S.A.; Pontecorvo, M.J.; Dean, R.A.; Demattos, R. Phase 3 solanezumab trials: Secondary outcomes in mild alzheimer’s disease patients. Alzheimer’s Dement. 2016, 12, 110–120. [Google Scholar] [CrossRef] [Green Version]
  95. Sevigny, J.; Chiao, P.; Bussière, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces aβ plaques in alzheimer’s disease. Nature 2016, 537, 50–56. [Google Scholar] [CrossRef]
  96. Novak, P.; Zilka, N.; Zilkova, M.; Kovacech, B.; Skrabana, R.; Ondrus, M.; Fialova, L.; Kontsekova, E.; Otto, M.; Novak, M. Aadvac1, an active immunotherapy for alzheimer’s disease and non alzheimer tauopathies: An overview of preclinical and clinical development. J. Prev. Alzheimer’s Dis. 2019, 6, 63–69. [Google Scholar]
  97. Theunis, C.; Crespo-Biel, N.; Gafner, V.; Pihlgren, M.; López-Deber, M.P.; Reis, P.; Hickman, D.T.; Adolfsson, O.; Chuard, N.; Ndao, D.M.; et al. Efficacy and safety of a liposome-based vaccine against protein tau, assessed in tau.P301l mice that model tauopathy. PLoS ONE 2013, 8, e72301. [Google Scholar] [CrossRef] [Green Version]
  98. Davies, P.; Maloney, A.J. Selective loss of central cholinergic neurons in alzheimer’s disease. Lancet 1976, 2, 1403. [Google Scholar] [CrossRef]
  99. Jack, C.R.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. Nia-aa research framework: Toward a biological definition of alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
  100. Pedrero-Prieto, C.M.; García-Carpintero, S.; Frontiñán-Rubio, J.; Llanos-González, E.; Aguilera García, C.; Alcaín, F.J.; Lindberg, I.; Durán-Prado, M.; Peinado, J.R.; Rabanal-Ruiz, Y. A comprehensive systematic review of csf proteins and peptides that define alzheimer’s disease. Clin. Proteom. 2020, 17, 21. [Google Scholar] [CrossRef]
  101. Villemagne, V.L.; Doré, V.; Burnham, S.C.; Masters, C.L.; Rowe, C.C. Imaging tau and amyloid-β proteinopathies in alzheimer disease and other conditions. Nat. Rev. Neurol. 2018, 14, 225–236. [Google Scholar] [CrossRef] [PubMed]
  102. Mattsson-Carlgren, N.; Andersson, E.; Janelidze, S.; Ossenkoppele, R.; Insel, P.; Strandberg, O.; Zetterberg, H.; Rosen, H.J.; Rabinovici, G.; Chai, X.; et al. Aβ deposition is associated with increases in soluble and phosphorylated tau that precede a positive tau pet in alzheimer’s disease. Sci. Adv. 2020, 6, eaaz2387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lombardi, G.; Crescioli, G.; Cavedo, E.; Lucenteforte, E.; Casazza, G.; Bellatorre, A.G.; Lista, C.; Costantino, G.; Frisoni, G.; Virgili, G.; et al. Structural magnetic resonance imaging for the early diagnosis of dementia due to alzheimer’s disease in people with mild cognitive impairment. Cochrane Database Syst. Rev. 2020, 3, CD009628. [Google Scholar] [CrossRef] [PubMed]
  104. Khosravi, M.; Peter, J.; Wintering, N.A.; Serruya, M.; Shamchi, S.P.; Werner, T.J.; Alavi, A.; Newberg, A.B. 18f-fdg is a superior indicator of cognitive performance compared to 18f-florbetapir in alzheimer’s disease and mild cognitive impairment evaluation: A global quantitative analysis. J. Alzheimer’s Dis. 2019, 70, 1197–1207. [Google Scholar] [CrossRef]
  105. Zetterberg, H.; Skillbäck, T.; Mattsson, N.; Trojanowski, J.Q.; Portelius, E.; Shaw, L.M.; Weiner, M.W.; Blennow, K.; Initiative, A.s.D.N. Association of cerebrospinal fluid neurofilament light concentration with alzheimer disease progression. JAMA Neurol. 2016, 73, 60–67. [Google Scholar] [CrossRef]
  106. Janelidze, S.; Hertze, J.; Zetterberg, H.; Landqvist Waldö, M.; Santillo, A.; Blennow, K.; Hansson, O. Cerebrospinal fluid neurogranin and ykl-40 as biomarkers of alzheimer’s disease. Ann. Clin. Transl. Neurol. 2016, 3, 12–20. [Google Scholar] [CrossRef] [Green Version]
  107. Jack, C.R.; Albert, M.S.; Knopman, D.S.; McKhann, G.M.; Sperling, R.A.; Carrillo, M.C.; Thies, B.; Phelps, C.H. Introduction to the recommendations from the national institute on aging-alzheimer’s association workgroups on diagnostic guidelines for alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 257–262. [Google Scholar] [CrossRef] [Green Version]
  108. McKhann, G.M.; Knopman, D.S.; Chertkow, H.; Hyman, B.T.; Jack, C.R.; Kawas, C.H.; Klunk, W.E.; Koroshetz, W.J.; Manly, J.J.; 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. Alzheimer’s Dement. 2011, 7, 263–269. [Google Scholar] [CrossRef] [Green Version]
  109. Albert, M.S.; DeKosky, S.T.; Dickson, D.; Dubois, B.; Feldman, H.H.; Fox, N.C.; Gamst, A.; Holtzman, D.M.; Jagust, W.J.; Petersen, R.C.; 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. Alzheimer’s Dement. 2011, 7, 270–279. [Google Scholar] [CrossRef] [Green Version]
  110. Sperling, R.A.; Aisen, P.S.; Beckett, L.A.; Bennett, D.A.; Craft, S.; Fagan, A.M.; Iwatsubo, T.; Jack, C.R.; Kaye, J.; Montine, T.J.; et al. Toward defining the preclinical stages of 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, 280–292. [Google Scholar] [CrossRef] [Green Version]
  111. Dubois, B.; Feldman, H.H.; Jacova, C.; Dekosky, S.T.; Barberger-Gateau, P.; Cummings, J.; Delacourte, A.; Galasko, D.; Gauthier, S.; Jicha, G.; et al. Research criteria for the diagnosis of alzheimer’s disease: Revising the nincds-adrda criteria. Lancet Neurol. 2007, 6, 734–746. [Google Scholar] [CrossRef]
  112. Dubois, B.; Hampel, H.; Feldman, H.H.; Scheltens, P.; Aisen, P.; Andrieu, S.; Bakardjian, H.; Benali, H.; Bertram, L.; Blennow, K.; et al. Preclinical alzheimer’s disease: Definition, natural history, and diagnostic criteria. Alzheimer’s Dement. 2016, 12, 292–323. [Google Scholar] [CrossRef] [PubMed]
  113. Hampel, H.; O’Bryant, S.E.; Molinuevo, J.L.; Zetterberg, H.; Masters, C.L.; Lista, S.; Kiddle, S.J.; Batrla, R.; Blennow, K. Blood-based biomarkers for alzheimer disease: Mapping the road to the clinic. Nat. Rev. Neurol. 2018, 14, 639–652. [Google Scholar] [CrossRef] [PubMed]
  114. Twohig, D.; Rodriguez-Vieitez, E.; Sando, S.B.; Berge, G.; Lauridsen, C.; Møller, I.; Grøntvedt, G.R.; Bråthen, G.; Patra, K.; Bu, G.; et al. The relevance of cerebrospinal fluid α-synuclein levels to sporadic and familial alzheimer’s disease. Acta Neuropathol. Commun. 2018, 6, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Olsson, B.; Lautner, R.; Andreasson, U.; Öhrfelt, A.; Portelius, E.; Bjerke, M.; Hölttä, M.; Rosén, C.; Olsson, C.; Strobel, G.; et al. Csf and blood biomarkers for the diagnosis of alzheimer’s disease: A systematic review and meta-analysis. Lancet Neurol. 2016, 15, 673–684. [Google Scholar] [CrossRef]
  116. Li, Q.X.; Villemagne, V.L.; Doecke, J.D.; Rembach, A.; Sarros, S.; Varghese, S.; McGlade, A.; Laughton, K.M.; Pertile, K.K.; Fowler, C.J.; et al. Alzheimer’s disease normative cerebrospinal fluid biomarkers validated in pet amyloid-β characterized subjects from the australian imaging, biomarkers and lifestyle (aibl) study. J. Alzheimer’s Dis. 2015, 48, 175–187. [Google Scholar] [CrossRef]
  117. Mattsson, N.; Palmqvist, S.; Stomrud, E.; Vogel, J.; Hansson, O. Staging β-amyloid pathology with amyloid positron emission tomography. JAMA Neurol. 2019, 76, 1319–1329. [Google Scholar] [CrossRef]
  118. Fagan, A.M.; Xiong, C.; Jasielec, M.S.; Bateman, R.J.; Goate, A.M.; Benzinger, T.L.; Ghetti, B.; Martins, R.N.; Masters, C.L.; Mayeux, R.; et al. Longitudinal change in csf biomarkers in autosomal-dominant alzheimer’s disease. Sci. Transl. Med. 2014, 6, 226ra230. [Google Scholar] [CrossRef] [Green Version]
  119. Lewczuk, P.; Matzen, A.; Blennow, K.; Parnetti, L.; Molinuevo, J.L.; Eusebi, P.; Kornhuber, J.; Morris, J.C.; Fagan, A.M. Cerebrospinal fluid aβ42/40 corresponds better than aβ42 to amyloid pet in alzheimer’s disease. J. Alzheimer’s Dis. 2017, 55, 813–822. [Google Scholar] [CrossRef] [Green Version]
  120. Doecke, J.D.; Rembach, A.; Villemagne, V.L.; Varghese, S.; Rainey-Smith, S.; Sarros, S.; Evered, L.A.; Fowler, C.J.; Pertile, K.K.; Rumble, R.L.; et al. Concordance between cerebrospinal fluid biomarkers with alzheimer’s disease pathology between three independent assay platforms. J. Alzheimer’s Dis. 2018, 61, 169–183. [Google Scholar] [CrossRef]
  121. Xia, D.; Watanabe, H.; Wu, B.; Lee, S.H.; Li, Y.; Tsvetkov, E.; Bolshakov, V.Y.; Shen, J.; Kelleher, R.J. Presenilin-1 knockin mice reveal loss-of-function mechanism for familial alzheimer’s disease. Neuron 2015, 85, 967–981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Selkoe, D.J.; Hardy, J. The amyloid hypothesis of alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef] [PubMed]
  123. Saido, T.C.; Iwatsubo, T.; Mann, D.M.; Shimada, H.; Ihara, Y.; Kawashima, S. Dominant and differential deposition of distinct beta-amyloid peptide species, a beta n3(pe), in senile plaques. Neuron 1995, 14, 457–466. [Google Scholar] [CrossRef] [Green Version]
  124. Portelius, E.; Bogdanovic, N.; Gustavsson, M.K.; Volkmann, I.; Brinkmalm, G.; Zetterberg, H.; Winblad, B.; Blennow, K. Mass spectrometric characterization of brain amyloid beta isoform signatures in familial and sporadic alzheimer’s disease. Acta Neuropathol. 2010, 120, 185–193. [Google Scholar] [CrossRef] [Green Version]
  125. Nussbaum, J.M.; Schilling, S.; Cynis, H.; Silva, A.; Swanson, E.; Wangsanut, T.; Tayler, K.; Wiltgen, B.; Hatami, A.; Rönicke, R.; et al. Prion-like behaviour and tau-dependent cytotoxicity of pyroglutamylated amyloid-β. Nature 2012, 485, 651–655. [Google Scholar] [CrossRef] [Green Version]
  126. Plant, L.D.; Boyle, J.P.; Smith, I.F.; Peers, C.; Pearson, H.A. The production of amyloid beta peptide is a critical requirement for the viability of central neurons. J. Neurosci. 2003, 23, 5531–5535. [Google Scholar] [CrossRef]
  127. Sjögren, M.; Gisslén, M.; Vanmechelen, E.; Blennow, K. Low cerebrospinal fluid beta-amyloid 42 in patients with acute bacterial meningitis and normalization after treatment. Neurosci. Lett. 2001, 314, 33–36. [Google Scholar] [CrossRef]
  128. Emmerling, M.R.; Morganti-Kossmann, M.C.; Kossmann, T.; Stahel, P.F.; Watson, M.D.; Evans, L.M.; Mehta, P.D.; Spiegel, K.; Kuo, Y.M.; Roher, A.E.; et al. Traumatic brain injury elevates the alzheimer’s amyloid peptide a beta 42 in human csf. A possible role for nerve cell injury. Ann. N. Y. Acad. Sci. 2000, 903, 118–122. [Google Scholar] [CrossRef]
  129. Mondello, S.; Buki, A.; Barzo, P.; Randall, J.; Provuncher, G.; Hanlon, D.; Wilson, D.; Kobeissy, F.; Jeromin, A. Csf and plasma amyloid-beta temporal profiles and relationships with neurological status and mortality after severe traumatic brain injury. Sci. Rep. 2014, 4, 6446. [Google Scholar] [CrossRef] [Green Version]
  130. Olsson, A.; Csajbok, L.; Ost, M.; Höglund, K.; Nylén, K.; Rosengren, L.; Nellgård, B.; Blennow, K. Marked increase of beta-amyloid(1-42) and amyloid precursor protein in ventricular cerebrospinal fluid after severe traumatic brain injury. J. Neurol. 2004, 251, 870–876. [Google Scholar] [CrossRef]
  131. Hortobágyi, T.; Wise, S.; Hunt, N.; Cary, N.; Djurovic, V.; Fegan-Earl, A.; Shorrock, K.; Rouse, D.; Al-Sarraj, S. Traumatic axonal damage in the brain can be detected using beta-app immunohistochemistry within 35 min after head injury to human adults. Neuropathol. Appl. Neurobiol. 2007, 33, 226–237. [Google Scholar] [CrossRef] [PubMed]
  132. Tsitsopoulos, P.P.; Marklund, N. Amyloid-beta peptides and tau protein as biomarkers in cerebrospinal and interstitial fluid following traumatic brain injury: A review of experimental and clinical studies. Front. Neurol. 2013, 4, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Schindler, S.E.; Li, Y.; Todd, K.W.; Herries, E.M.; Henson, R.L.; Gray, J.D.; Wang, G.; Graham, D.L.; Shaw, L.M.; Trojanowski, J.Q.; et al. Emerging cerebrospinal fluid biomarkers in autosomal dominant alzheimer’s disease. Alzheimer’s Dement. 2019, 15, 655–665. [Google Scholar] [CrossRef] [PubMed]
  134. Henson, R.L.; Doran, E.; Christian, B.T.; Handen, B.L.; Klunk, W.E.; Lai, F.; Lee, J.H.; Rosas, H.D.; Schupf, N.; Zaman, S.H.; et al. Cerebrospinal fluid biomarkers of alzheimer’s disease in a cohort of adults with down syndrome. Alzheimer’s Dement. 2020, 12, e12057. [Google Scholar] [CrossRef] [PubMed]
  135. Mo, J.A.; Lim, J.H.; Sul, A.R.; Lee, M.; Youn, Y.C.; Kim, H.J. Cerebrospinal fluid β-amyloid1-42 levels in the differential diagnosis of alzheimer’s disease--systematic review and meta-analysis. PLoS ONE 2015, 10, e0116802. [Google Scholar] [CrossRef]
  136. Englund, H.; Annerén, G.; Gustafsson, J.; Wester, U.; Wiltfang, J.; Lannfelt, L.; Blennow, K.; Höglund, K. Increase in beta-amyloid levels in cerebrospinal fluid of children with down syndrome. Dement. Geriatr. Cogn. Disord. 2007, 24, 369–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Peskind, E.R.; Riekse, R.; Quinn, J.F.; Kaye, J.; Clark, C.M.; Farlow, M.R.; Decarli, C.; Chabal, C.; Vavrek, D.; Raskind, M.A.; et al. Safety and acceptability of the research lumbar puncture. Alzheimer Dis. Assoc. Disord. 2005, 19, 220–225. [Google Scholar] [CrossRef]
  138. Palmert, M.R.; Podlisny, M.B.; Witker, D.S.; Oltersdorf, T.; Younkin, L.H.; Selkoe, D.J.; Younkin, S.G. The beta-amyloid protein precursor of alzheimer disease has soluble derivatives found in human brain and cerebrospinal fluid. Proc. Natl. Acad. Sci. USA 1989, 86, 6338–6342. [Google Scholar] [CrossRef] [Green Version]
  139. Ghiso, J.; Tagliavini, F.; Timmers, W.F.; Frangione, B. Alzheimer’s disease amyloid precursor protein is present in senile plaques and cerebrospinal fluid: Immunohistochemical and biochemical characterization. Biochem. Biophys. Res. Commun. 1989, 163, 430–437. [Google Scholar] [CrossRef]
  140. Palmert, M.R.; Siedlak, S.L.; Podlisny, M.B.; Greenberg, B.; Shelton, E.R.; Chan, H.W.; Usiak, M.; Selkoe, D.J.; Perry, G.; Younkin, S.G. Soluble derivatives of the beta amyloid protein precursor of alzheimer’s disease are labeled by antisera to the beta amyloid protein. Biochem. Biophys. Res. Commun. 1989, 165, 182–188. [Google Scholar] [CrossRef]
  141. Chong, J.K.; Miller, B.E.; Ghanbari, H.A. Detection of amyloid beta protein precursor immunoreactivity in normal and alzheimer’s disease cerebrospinal fluid. Life Sci. 1990, 47, 1163–1171. [Google Scholar] [CrossRef]
  142. Van Nostrand, W.E.; Wagner, S.L.; Shankle, W.R.; Farrow, J.S.; Dick, M.; Rozemuller, J.M.; Kuiper, M.A.; Wolters, E.C.; Zimmerman, J.; Cotman, C.W. Decreased levels of soluble amyloid beta-protein precursor in cerebrospinal fluid of live alzheimer disease patients. Proc. Natl. Acad. Sci. USA 1992, 89, 2551–2555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Urakami, K.; Takahashi, K.; Okada, A.; Oshima, T.; Adachi, Y.; Nakamura, S.; Kitaguchi, N.; Tokushima, Y.; Yamamoto, S.; Tanaka, S. Clinical course and csf amyloid beta protein precursor having the site of application of the protease inhibitor (appi) levels in patients with dementia of the alzheimer type. Dementia 1993, 4, 59–60. [Google Scholar]
  144. Vigo-Pelfrey, C.; Lee, D.; Keim, P.; Lieberburg, I.; Schenk, D.B. Characterization of beta-amyloid peptide from human cerebrospinal fluid. J. Neurochem. 1993, 61, 1965–1968. [Google Scholar] [CrossRef]
  145. van Gool, W.A.; Schenk, D.B.; Bolhuis, P.A. Concentrations of amyloid-beta protein in cerebrospinal fluid increase with age in patients free from neurodegenerative disease. Neurosci. Lett. 1994, 172, 122–124. [Google Scholar] [CrossRef]
  146. Tabaton, M.; Nunzi, M.G.; Xue, R.; Usiak, M.; Autilio-Gambetti, L.; Gambetti, P. Soluble amyloid beta-protein is a marker of alzheimer amyloid in brain but not in cerebrospinal fluid. Biochem. Biophys. Res. Commun. 1994, 200, 1598–1603. [Google Scholar] [CrossRef]
  147. Jensen, M.; Schröder, J.; Blomberg, M.; Engvall, B.; Pantel, J.; Ida, N.; Basun, H.; Wahlund, L.O.; Werle, E.; Jauss, M.; et al. Cerebrospinal fluid a beta42 is increased early in sporadic alzheimer’s disease and declines with disease progression. Ann. Neurol. 1999, 45, 504–511. [Google Scholar] [CrossRef]
  148. Fagan, A.M.; Roe, C.M.; Xiong, C.; Mintun, M.A.; Morris, J.C.; Holtzman, D.M. Cerebrospinal fluid tau/beta-amyloid(42) ratio as a prediction of cognitive decline in nondemented older adults. Arch. Neurol. 2007, 64, 343–349. [Google Scholar] [CrossRef]
  149. Fagan, A.M.; Perrin, R.J. Upcoming candidate cerebrospinal fluid biomarkers of alzheimer’s disease. Biomark. Med. 2012, 6, 455–476. [Google Scholar] [CrossRef] [Green Version]
  150. Hansson, O.; Lehmann, S.; Otto, M.; Zetterberg, H.; Lewczuk, P. Advantages and disadvantages of the use of the csf amyloid β (aβ) 42/40 ratio in the diagnosis of alzheimer’s disease. Alzheimer’s Res. Ther. 2019, 11, 34. [Google Scholar] [CrossRef]
  151. Holtzman, D.M. Csf biomarkers for alzheimer’s disease: Current utility and potential future use. Neurobiol. Aging 2011, 32 (Suppl. 1), S4–S9. [Google Scholar] [CrossRef] [Green Version]
  152. Dumurgier, J.; Gabelle, A.; Vercruysse, O.; Bombois, S.; Laplanche, J.L.; Peoc’h, K.; Schraen, S.; Sablonnière, B.; Pasquier, F.; Touchon, J.; et al. Exacerbated csf abnormalities in younger patients with alzheimer’s disease. Neurobiol. Dis. 2013, 54, 486–491. [Google Scholar] [CrossRef] [PubMed]
  153. Reijn, T.S.; Rikkert, M.O.; van Geel, W.J.; de Jong, D.; Verbeek, M.M. Diagnostic accuracy of elisa and xmap technology for analysis of amyloid beta(42) and tau proteins. Clin. Chem. 2007, 53, 859–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Mattsson, N.; Zetterberg, H.; Hansson, O.; Andreasen, N.; Parnetti, L.; Jonsson, M.; Herukka, S.K.; van der Flier, W.M.; Blankenstein, M.A.; Ewers, M.; et al. Csf biomarkers and incipient alzheimer disease in patients with mild cognitive impairment. JAMA 2009, 302, 385–393. [Google Scholar] [CrossRef] [PubMed]
  155. Kapaki, E.N.; Paraskevas, G.P.; Tzerakis, N.G.; Sfagos, C.; Seretis, A.; Kararizou, E.; Vassilopoulos, D. Cerebrospinal fluid tau, phospho-tau181 and beta-amyloid1-42 in idiopathic normal pressure hydrocephalus: A discrimination from alzheimer’s disease. Eur. J. Neurol. 2007, 14, 168–173. [Google Scholar] [CrossRef] [PubMed]
  156. Kapaki, E.; Liappas, I.; Paraskevas, G.P.; Theotoka, I.; Rabavilas, A. The diagnostic value of tau protein, beta-amyloid (1-42) and their ratio for the discrimination of alcohol-related cognitive disorders from alzheimer’s disease in the early stages. Int. J. Geriatr. Psychiatry 2005, 20, 722–729. [Google Scholar] [CrossRef]
  157. Ewers, M.; Mattsson, N.; Minthon, L.; Molinuevo, J.L.; Antonell, A.; Popp, J.; Jessen, F.; Herukka, S.K.; Soininen, H.; Maetzler, W.; et al. Csf biomarkers for the differential diagnosis of alzheimer’s disease: A large-scale international multicenter study. Alzheimer’s Dement. 2015, 11, 1306–1315. [Google Scholar] [CrossRef]
  158. Selnes, P.; Blennow, K.; Zetterberg, H.; Grambaite, R.; Rosengren, L.; Johnsen, L.; Stenset, V.; Fladby, T. Effects of cerebrovascular disease on amyloid precursor protein metabolites in cerebrospinal fluid. Cerebrospinal Fluid Res. 2010, 7, 10. [Google Scholar] [CrossRef] [Green Version]
  159. Vos, S.J.; van Rossum, I.A.; Verhey, F.; Knol, D.L.; Soininen, H.; Wahlund, L.O.; Hampel, H.; Tsolaki, M.; Minthon, L.; Frisoni, G.B.; et al. Prediction of alzheimer disease in subjects with amnestic and nonamnestic mci. Neurology 2013, 80, 1124–1132. [Google Scholar] [CrossRef]
  160. Hampel, H.; Teipel, S.J.; Fuchsberger, T.; Andreasen, N.; Wiltfang, J.; Otto, M.; Shen, Y.; Dodel, R.; Du, Y.; Farlow, M.; et al. Value of csf beta-amyloid1-42 and tau as predictors of alzheimer’s disease in patients with mild cognitive impairment. Mol. Psychiatry 2004, 9, 705–710. [Google Scholar] [CrossRef] [Green Version]
  161. Perneczky, R.; Tsolakidou, A.; Arnold, A.; Diehl-Schmid, J.; Grimmer, T.; Förstl, H.; Kurz, A.; Alexopoulos, P. Csf soluble amyloid precursor proteins in the diagnosis of incipient alzheimer disease. Neurology 2011, 77, 35–38. [Google Scholar] [CrossRef] [PubMed]
  162. Lewczuk, P.; Kornhuber, J.; Vanderstichele, H.; Vanmechelen, E.; Esselmann, H.; Bibl, M.; Wolf, S.; Otto, M.; Reulbach, U.; Kölsch, H.; et al. Multiplexed quantification of dementia biomarkers in the csf of patients with early dementias and mci: A multicenter study. Neurobiol. Aging 2008, 29, 812–818. [Google Scholar] [CrossRef] [PubMed]
  163. Bibl, M.; Gallus, M.; Welge, V.; Esselmann, H.; Wolf, S.; Rüther, E.; Wiltfang, J. Cerebrospinal fluid amyloid-β 2-42 is decreased in alzheimer’s, but not in frontotemporal dementia. J. Neural Transm. 2012, 119, 805–813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Lewczuk, P.; Lelental, N.; Spitzer, P.; Maler, J.M.; Kornhuber, J. Amyloid-β 42/40 cerebrospinal fluid concentration ratio in the diagnostics of alzheimer’s disease: Validation of two novel assays. J. Alzheimer’s Dis. 2015, 43, 183–191. [Google Scholar] [CrossRef]
  165. Struyfs, H.; Van Broeck, B.; Timmers, M.; Fransen, E.; Sleegers, K.; Van Broeckhoven, C.; De Deyn, P.P.; Streffer, J.R.; Mercken, M.; Engelborghs, S. Diagnostic accuracy of cerebrospinal fluid amyloid-β isoforms for early and differential dementia diagnosis. J. Alzheimer’s Dis. 2015, 45, 813–822. [Google Scholar] [CrossRef] [Green Version]
  166. Hansson, O.; Zetterberg, H.; Buchhave, P.; Andreasson, U.; Londos, E.; Minthon, L.; Blennow, K. Prediction of alzheimer’s disease using the csf abeta42/abeta40 ratio in patients with mild cognitive impairment. Dement. Geriatr. Cogn. Disord. 2007, 23, 316–320. [Google Scholar] [CrossRef]
  167. Janelidze, S.; Zetterberg, H.; Mattsson, N.; Palmqvist, S.; Vanderstichele, H.; Lindberg, O.; van Westen, D.; Stomrud, E.; Minthon, L.; Blennow, K.; et al. Csf aβ42/aβ40 and aβ42/aβ38 ratios: Better diagnostic markers of alzheimer disease. Ann. Clin. Transl. Neurol. 2016, 3, 154–165. [Google Scholar] [CrossRef] [Green Version]
  168. Fukumoto, H.; Tennis, M.; Locascio, J.J.; Hyman, B.T.; Growdon, J.H.; Irizarry, M.C. Age but not diagnosis is the main predictor of plasma amyloid beta-protein levels. Arch. Neurol. 2003, 60, 958–964. [Google Scholar] [CrossRef] [Green Version]
  169. Rissin, D.M.; Kan, C.W.; Campbell, T.G.; Howes, S.C.; Fournier, D.R.; Song, L.; Piech, T.; Patel, P.P.; Chang, L.; Rivnak, A.J.; et al. Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat. Biotechnol. 2010, 28, 595–599. [Google Scholar] [CrossRef] [Green Version]
  170. Janelidze, S.; Stomrud, E.; Palmqvist, S.; Zetterberg, H.; van Westen, D.; Jeromin, A.; Song, L.; Hanlon, D.; Tan Hehir, C.A.; Baker, D.; et al. Plasma β-amyloid in alzheimer’s disease and vascular disease. Sci. Rep. 2016, 6, 26801. [Google Scholar] [CrossRef]
  171. Zetterberg, H.; Mörtberg, E.; Song, L.; Chang, L.; Provuncher, G.K.; Patel, P.P.; Ferrell, E.; Fournier, D.R.; Kan, C.W.; Campbell, T.G.; et al. Hypoxia due to cardiac arrest induces a time-dependent increase in serum amyloid β levels in humans. PLoS ONE 2011, 6, e28263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Yang, S.Y.; Chiu, M.J.; Chen, T.F.; Horng, H.E. Detection of plasma biomarkers using immunomagnetic reduction: A promising method for the early diagnosis of alzheimer’s disease. Neurol. Ther. 2017, 6, 37–56. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, L.S.; Leung, Y.Y.; Chang, S.K.; Leight, S.; Knapik-Czajka, M.; Baek, Y.; Shaw, L.M.; Lee, V.M.; Trojanowski, J.Q.; Clark, C.M. Comparison of xmap and elisa assays for detecting cerebrospinal fluid biomarkers of alzheimer’s disease. J. Alzheimer’s Dis. 2012, 31, 439–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Pan, C.; Korff, A.; Galasko, D.; Ginghina, C.; Peskind, E.; Li, G.; Quinn, J.; Montine, T.J.; Cain, K.; Shi, M.; et al. Diagnostic values of cerebrospinal fluid t-tau and aβ42 using meso scale discovery assays for alzheimer’s disease. J. Alzheimer’s Dis. 2015, 45, 709–719. [Google Scholar] [CrossRef] [Green Version]
  175. Nakamura, A.; Kaneko, N.; Villemagne, V.L.; Kato, T.; Doecke, J.; Doré, V.; Fowler, C.; Li, Q.X.; Martins, R.; Rowe, C.; et al. High performance plasma amyloid-β biomarkers for alzheimer’s disease. Nature 2018, 554, 249–254. [Google Scholar] [CrossRef]
  176. Schindler, S.E.; Bollinger, J.G.; Ovod, V.; Mawuenyega, K.G.; Li, Y.; Gordon, B.A.; Holtzman, D.M.; Morris, J.C.; Benzinger, T.L.S.; Xiong, C.; et al. High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology 2019, 93, e1647–e1659. [Google Scholar] [CrossRef]
  177. Verberk, I.M.W.; Slot, R.E.; Verfaillie, S.C.J.; Heijst, H.; Prins, N.D.; van Berckel, B.N.M.; Scheltens, P.; Teunissen, C.E.; van der Flier, W.M. Plasma amyloid as prescreener for the earliest alzheimer pathological changes. Ann. Neurol. 2018, 84, 648–658. [Google Scholar] [CrossRef]
  178. Kaneko, N.; Nakamura, A.; Washimi, Y.; Kato, T.; Sakurai, T.; Arahata, Y.; Bundo, M.; Takeda, A.; Niida, S.; Ito, K.; et al. Novel plasma biomarker surrogating cerebral amyloid deposition. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2014, 90, 353–364. [Google Scholar] [CrossRef] [Green Version]
  179. Ovod, V.; Ramsey, K.N.; Mawuenyega, K.G.; Bollinger, J.G.; Hicks, T.; Schneider, T.; Sullivan, M.; Paumier, K.; Holtzman, D.M.; Morris, J.C.; et al. Amyloid β concentrations and stable isotope labeling kinetics of human plasma specific to central nervous system amyloidosis. Alzheimer’s Dement. 2017, 13, 841–849. [Google Scholar] [CrossRef]
  180. Pannee, J.; Törnqvist, U.; Westerlund, A.; Ingelsson, M.; Lannfelt, L.; Brinkmalm, G.; Persson, R.; Gobom, J.; Svensson, J.; Johansson, P.; 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. [Google Scholar] [CrossRef]
  181. Li, Q.X.; Berndt, M.C.; Bush, A.I.; Rumble, B.; Mackenzie, I.; Friedhuber, A.; Beyreuther, K.; Masters, C.L. Membrane-associated forms of the beta a4 amyloid protein precursor of alzheimer’s disease in human platelet and brain: Surface expression on the activated human platelet. Blood 1994, 84, 133–142. [Google Scholar] [CrossRef] [PubMed]
  182. Palmqvist, S.; Insel, P.S.; Stomrud, E.; Janelidze, S.; Zetterberg, H.; Brix, B.; Eichenlaub, U.; Dage, J.L.; Chai, X.; Blennow, K.; et al. Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in alzheimer’s disease. EMBO Mol. Med. 2019, 11, e11170. [Google Scholar] [CrossRef]
  183. Augustinack, J.C.; Schneider, A.; Mandelkow, E.M.; Hyman, B.T. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in alzheimer’s disease. Acta Neuropathol. 2002, 103, 26–35. [Google Scholar] [CrossRef] [PubMed]
  184. Haroutunian, V.; Davies, P.; Vianna, C.; Buxbaum, J.D.; Purohit, D.P. Tau protein abnormalities associated with the progression of alzheimer disease type dementia. Neurobiol. Aging 2007, 28, 1–7. [Google Scholar] [CrossRef] [PubMed]
  185. Zetterberg, H.; Bendlin, B.B. Biomarkers for alzheimer’s disease-preparing for a new era of disease-modifying therapies. Mol. Psychiatry 2020, 1–13. [Google Scholar] [CrossRef] [PubMed]
  186. Fossati, S.; Ramos Cejudo, J.; Debure, L.; Pirraglia, E.; Sone, J.Y.; Li, Y.; Chen, J.; Butler, T.; Zetterberg, H.; Blennow, K.; et al. Plasma tau complements csf tau and p-tau in the diagnosis of alzheimer’s disease. Alzheimer’s Dement. 2019, 11, 483–492. [Google Scholar] [CrossRef] [PubMed]
  187. Vandermeeren, M.; Mercken, M.; Vanmechelen, E.; Six, J.; van de Voorde, A.; Martin, J.J.; Cras, P. Detection of tau proteins in normal and alzheimer’s disease cerebrospinal fluid with a sensitive sandwich enzyme-linked immunosorbent assay. J. Neurochem. 1993, 61, 1828–1834. [Google Scholar] [CrossRef]
  188. Kandimalla, R.J.; Prabhakar, S.; Bk, B.; Wani, W.Y.; Sharma, D.R.; Grover, V.K.; Bhardwaj, N.; Jain, K.; Gill, K.D. Cerebrospinal fluid profile of amyloid β42 (aβ42), htau and ubiquitin in north indian alzheimer’s disease patients. Neurosci. Lett. 2011, 487, 134–138. [Google Scholar] [CrossRef]
  189. Kandimalla, R.J.; Prabhakar, S.; Binukumar, B.K.; Wani, W.Y.; Gupta, N.; Sharma, D.R.; Sunkaria, A.; Grover, V.K.; Bhardwaj, N.; Jain, K.; et al. Apo-eε4 allele in conjunction with aβ42 and tau in csf: Biomarker for alzheimer’s disease. Curr. Alzheimer Res. 2011, 8, 187–196. [Google Scholar] [CrossRef]
  190. Tapiola, T.; Alafuzoff, I.; Herukka, S.K.; Parkkinen, L.; Hartikainen, P.; Soininen, H.; Pirttilä, T. Cerebrospinal fluid {beta}-amyloid 42 and tau proteins as biomarkers of alzheimer-type pathologic changes in the brain. Arch Neurol. 2009, 66, 382–389. [Google Scholar] [CrossRef] [Green Version]
  191. Haense, C.; Buerger, K.; Kalbe, E.; Drzezga, A.; Teipel, S.J.; Markiewicz, P.; Herholz, K.; Heiss, W.D.; Hampel, H. Csf total and phosphorylated tau protein, regional glucose metabolism and dementia severity in alzheimer’s disease. Eur. J. Neurol. 2008, 15, 1155–1162. [Google Scholar] [CrossRef] [PubMed]
  192. Hampel, H.; Goernitz, A.; Buerger, K. Advances in the development of biomarkers for alzheimer’s disease: From csf total tau and abeta(1-42) proteins to phosphorylated tau protein. Brain Res. Bull. 2003, 61, 243–253. [Google Scholar] [CrossRef]
  193. Gordon, B.A.; Blazey, T.M.; Christensen, J.; Dincer, A.; Flores, S.; Keefe, S.; Chen, C.; Su, Y.; McDade, E.M.; Wang, G.; et al. Tau pet in autosomal dominant alzheimer’s disease: Relationship with cognition, dementia and other biomarkers. Brain 2019, 142, 1063–1076. [Google Scholar] [CrossRef]
  194. Quiroz, Y.T.; Sperling, R.A.; Norton, D.J.; Baena, A.; Arboleda-Velasquez, J.F.; Cosio, D.; Schultz, A.; Lapoint, M.; Guzman-Velez, E.; Miller, J.B.; et al. Association between amyloid and tau accumulation in young adults with autosomal dominant alzheimer disease. JAMA Neurol. 2018, 75, 548–556. [Google Scholar] [CrossRef] [PubMed]
  195. Fleisher, A.S.; Chen, K.; Quiroz, Y.T.; Jakimovich, L.J.; Gutierrez Gomez, M.; Langois, C.M.; Langbaum, J.B.; Roontiva, A.; Thiyyagura, P.; Lee, W.; et al. Associations between biomarkers and age in the presenilin 1 e280a autosomal dominant alzheimer disease kindred: A cross-sectional study. JAMA Neurol. 2015, 72, 316–324. [Google Scholar] [CrossRef] [PubMed]
  196. Toledo, J.B.; Xie, S.X.; Trojanowski, J.Q.; Shaw, L.M. Longitudinal change in csf tau and aβ biomarkers for up to 48 months in adni. Acta Neuropathol. 2013, 126, 659–670. [Google Scholar] [CrossRef] [Green Version]
  197. McDade, E.; Wang, G.; Gordon, B.A.; Hassenstab, J.; Benzinger, T.L.S.; Buckles, V.; Fagan, A.M.; Holtzman, D.M.; Cairns, N.J.; Goate, A.M.; et al. Longitudinal cognitive and biomarker changes in dominantly inherited alzheimer disease. Neurology 2018, 91, e1295–e1306. [Google Scholar] [CrossRef]
  198. Fitzpatrick, A.W.P.; Falcon, B.; He, S.; Murzin, A.G.; Murshudov, G.; Garringer, H.J.; Crowther, R.A.; Ghetti, B.; Goedert, M.; Scheres, S.H.W. Cryo-em structures of tau filaments from alzheimer’s disease. Nature 2017, 547, 185–190. [Google Scholar] [CrossRef] [Green Version]
  199. Falcon, B.; Zhang, W.; Schweighauser, M.; Murzin, A.G.; Vidal, R.; Garringer, H.J.; Ghetti, B.; Scheres, S.H.W.; Goedert, M. Tau filaments from multiple cases of sporadic and inherited alzheimer’s disease adopt a common fold. Acta Neuropathol. 2018, 136, 699–708. [Google Scholar] [CrossRef] [Green Version]
  200. Zhang, W.; Falcon, B.; Murzin, A.G.; Fan, J.; Crowther, R.A.; Goedert, M.; Scheres, S.H. Heparin-induced tau filaments are polymorphic and differ from those in alzheimer’s and pick’s diseases. Elife 2019, 8, e43584. [Google Scholar] [CrossRef]
  201. Arakhamia, T.; Lee, C.E.; Carlomagno, Y.; Duong, D.M.; Kundinger, S.R.; Wang, K.; Williams, D.; DeTure, M.; Dickson, D.W.; Cook, C.N.; et al. Posttranslational modifications mediate the structural diversity of tauopathy strains. Cell 2020, 180, 633–644. [Google Scholar] [CrossRef] [PubMed]
  202. Jicha, G.A.; Bowser, R.; Kazam, I.G.; Davies, P. Alz-50 and mc-1, a new monoclonal antibody raised to paired helical filaments, recognize conformational epitopes on recombinant tau. J. Neurosci. Res. 1997, 48, 128–132. [Google Scholar] [CrossRef]
  203. Jicha, G.A.; Lane, E.; Vincent, I.; Otvos, L.; Hoffmann, R.; Davies, P. A conformation- and phosphorylation-dependent antibody recognizing the paired helical filaments of alzheimer’s disease. J. Neurochem. 1997, 69, 2087–2095. [Google Scholar] [CrossRef] [PubMed]
  204. Weaver, C.L.; Espinoza, M.; Kress, Y.; Davies, P. Conformational change as one of the earliest alterations of tau in alzheimer’s disease. Neurobiol. Aging 2000, 21, 719–727. [Google Scholar] [CrossRef]
  205. Cicognola, C.; Brinkmalm, G.; Wahlgren, J.; Portelius, E.; Gobom, J.; Cullen, N.C.; Hansson, O.; Parnetti, L.; Constantinescu, R.; Wildsmith, K.; et al. Novel tau fragments in cerebrospinal fluid: Relation to tangle pathology and cognitive decline in alzheimer’s disease. Acta Neuropathol. 2019, 137, 279–296. [Google Scholar] [CrossRef] [Green Version]
  206. Amadoro, G.; Corsetti, V.; Sancesario, G.M.; Lubrano, A.; Melchiorri, G.; Bernardini, S.; Calissano, P.; Sancesario, G. Cerebrospinal fluid levels of a 20-22 kda nh2 fragment of human tau provide a novel neuronal injury biomarker in alzheimer’s disease and other dementias. J. Alzheimer’s Dis. 2014, 42, 211–226. [Google Scholar] [CrossRef]
  207. Blennow, K.; Chen, C.; Cicognola, C.; Wildsmith, K.R.; Manser, P.T.; Bohorquez, S.M.S.; Zhang, Z.; Xie, B.; Peng, J.; Hansson, O.; et al. Cerebrospinal fluid tau fragment correlates with tau pet: A candidate biomarker for tangle pathology. Brain 2020, 143, 650–660. [Google Scholar] [CrossRef]
  208. Quinn, J.P.; Corbett, N.J.; Kellett, K.A.B.; Hooper, N.M. Tau proteolysis in the pathogenesis of tauopathies: Neurotoxic fragments and novel biomarkers. J. Alzheimer’s Dis. 2018, 63, 13–33. [Google Scholar] [CrossRef] [Green Version]
  209. Maia, L.F.; Kaeser, S.A.; Reichwald, J.; Hruscha, M.; Martus, P.; Staufenbiel, M.; Jucker, M. Changes in amyloid-β and tau in the cerebrospinal fluid of transgenic mice overexpressing amyloid precursor protein. Sci. Transl. Med. 2013, 5, 194re192. [Google Scholar] [CrossRef]
  210. Sato, C.; Barthélemy, N.R.; Mawuenyega, K.G.; Patterson, B.W.; Gordon, B.A.; Jockel-Balsarotti, J.; Sullivan, M.; Crisp, M.J.; Kasten, T.; Kirmess, K.M.; et al. Tau kinetics in neurons and the human central nervous system. Neuron 2018, 97, 1284–1298. [Google Scholar] [CrossRef] [Green Version]
  211. Buerger, K.; Ewers, M.; Pirttilä, T.; Zinkowski, R.; Alafuzoff, I.; Teipel, S.J.; DeBernardis, J.; Kerkman, D.; McCulloch, C.; Soininen, H.; et al. Csf phosphorylated tau protein correlates with neocortical neurofibrillary pathology in alzheimer’s disease. Brain 2006, 129, 3035–3041. [Google Scholar] [CrossRef] [PubMed]
  212. Molinuevo, J.L.; Ayton, S.; Batrla, R.; Bednar, M.M.; Bittner, T.; Cummings, J.; Fagan, A.M.; Hampel, H.; Mielke, M.M.; Mikulskis, A.; et al. Current state of alzheimer’s fluid biomarkers. Acta Neuropathol. 2018, 136, 821–853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Blennow, K.; Hampel, H.; Weiner, M.; Zetterberg, H. Cerebrospinal fluid and plasma biomarkers in alzheimer disease. Nat. Rev. Neurol. 2010, 6, 131–144. [Google Scholar] [CrossRef] [PubMed]
  214. Lleó, A.; Irwin, D.J.; Illán-Gala, I.; McMillan, C.T.; Wolk, D.A.; Lee, E.B.; Van Deerlin, V.M.; Shaw, L.M.; Trojanowski, J.Q.; Grossman, M. A 2-step cerebrospinal algorithm for the selection of frontotemporal lobar degeneration subtypes. JAMA Neurol. 2018, 75, 738–745. [Google Scholar] [CrossRef] [Green Version]
  215. Schoonenboom, N.S.; Reesink, F.E.; Verwey, N.A.; Kester, M.I.; Teunissen, C.E.; van de Ven, P.M.; Pijnenburg, Y.A.; Blankenstein, M.A.; Rozemuller, A.J.; Scheltens, P.; et al. Cerebrospinal fluid markers for differential dementia diagnosis in a large memory clinic cohort. Neurology 2012, 78, 47–54. [Google Scholar] [CrossRef] [Green Version]
  216. Buchhave, P.; Minthon, L.; Zetterberg, H.; Wallin, A.K.; Blennow, K.; Hansson, O. Cerebrospinal fluid levels of β-amyloid 1-42, but not of tau, are fully changed already 5 to 10 years before the onset of alzheimer dementia. Arch. Gen. Psychiatry 2012, 69, 98–106. [Google Scholar] [CrossRef]
  217. Vos, S.J.; Xiong, C.; Visser, P.J.; Jasielec, M.S.; Hassenstab, J.; Grant, E.A.; Cairns, N.J.; Morris, J.C.; Holtzman, D.M.; Fagan, A.M. Preclinical alzheimer’s disease and its outcome: A longitudinal cohort study. Lancet Neurol. 2013, 12, 957–965. [Google Scholar] [CrossRef] [Green Version]
  218. Hansson, O.; Mikulskis, A.; Fagan, A.M.; Teunissen, C.; Zetterberg, H.; Vanderstichele, H.; Molinuevo, J.L.; Shaw, L.M.; Vandijck, M.; Verbeek, M.M.; et al. The impact of preanalytical variables on measuring cerebrospinal fluid biomarkers for alzheimer’s disease diagnosis: A review. Alzheimer’s Dement. 2018, 14, 1313–1333. [Google Scholar] [CrossRef] [Green Version]
  219. Shaw, L.M.; Arias, J.; Blennow, K.; Galasko, D.; Molinuevo, J.L.; Salloway, S.; Schindler, S.; Carrillo, M.C.; Hendrix, J.A.; Ross, A.; et al. Appropriate use criteria for lumbar puncture and cerebrospinal fluid testing in the diagnosis of alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 1505–1521. [Google Scholar] [CrossRef]
  220. Janelidze, S.; Stomrud, E.; Smith, R.; Palmqvist, S.; Mattsson, N.; Airey, D.C.; Proctor, N.K.; Chai, X.; Shcherbinin, S.; Sims, J.R.; et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of alzheimer’s disease. Nat. Commun. 2020, 11, 1683. [Google Scholar] [CrossRef] [Green Version]
  221. Hampel, H.; Buerger, K.; Zinkowski, R.; Teipel, S.J.; Goernitz, A.; Andreasen, N.; Sjoegren, M.; DeBernardis, J.; Kerkman, D.; Ishiguro, K.; et al. Measurement of phosphorylated tau epitopes in the differential diagnosis of alzheimer disease: A comparative cerebrospinal fluid study. Arch. Gen. Psychiatry 2004, 61, 95–102. [Google Scholar] [CrossRef] [Green Version]
  222. Spiegel, J.; Pirraglia, E.; Osorio, R.S.; Glodzik, L.; Li, Y.; Tsui, W.; Saint Louis, L.A.; Randall, C.; Butler, T.; Xu, J.; et al. Greater specificity for cerebrospinal fluid p-tau231 over p-tau181 in the differentiation of healthy controls from alzheimer’s disease. J. Alzheimer’s Dis. 2016, 49, 93–100. [Google Scholar] [CrossRef] [Green Version]
  223. Buerger, K.; Alafuzoff, I.; Ewers, M.; Pirttilä, T.; Zinkowski, R.; Hampel, H. No correlation between csf tau protein phosphorylated at threonine 181 with neocortical neurofibrillary pathology in alzheimer’s disease. Brain 2007, 130, e82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Braak, H.; Braak, E. On areas of transition between entorhinal allocortex and temporal isocortex in the human brain. Normal morphology and lamina-specific pathology in alzheimer’s disease. Acta Neuropathol. 1985, 68, 325–332. [Google Scholar] [CrossRef] [PubMed]
  225. Barthélemy, N.R.; Bateman, R.J.; Hirtz, C.; Marin, P.; Becher, F.; Sato, C.; Gabelle, A.; Lehmann, S. Cerebrospinal fluid phospho-tau t217 outperforms t181 as a biomarker for the differential diagnosis of alzheimer’s disease and pet amyloid-positive patient identification. Alzheimer’s Res. Ther. 2020, 12, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Barthélemy, N.R.; Li, Y.; Joseph-Mathurin, N.; Gordon, B.A.; Hassenstab, J.; Benzinger, T.L.S.; Buckles, V.; Fagan, A.M.; Perrin, R.J.; Goate, A.M.; et al. A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited alzheimer’s disease. Nat. Med. 2020, 26, 398–407. [Google Scholar] [CrossRef]
  227. Chen, Z.; Mengel, D.; Keshavan, A.; Rissman, R.A.; Billinton, A.; Perkinton, M.; Percival-Alwyn, J.; Schultz, A.; Properzi, M.; Johnson, K.; et al. Learnings about the complexity of extracellular tau aid development of a blood-based screen for alzheimer’s disease. Alzheimer’s Dement. 2019, 15, 487–496. [Google Scholar] [CrossRef] [PubMed]
  228. Ramos-Cejudo, J.; Wisniewski, T.; Marmar, C.; Zetterberg, H.; Blennow, K.; de Leon, M.J.; Fossati, S. Traumatic brain injury and alzheimer’s disease: The cerebrovascular link. EBioMedicine 2018, 28, 21–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Mendez, M.F. What is the relationship of traumatic brain injury to dementia? J. Alzheimer’s Dis. 2017, 57, 667–681. [Google Scholar] [CrossRef]
  230. Blennow, K.; Brody, D.L.; Kochanek, P.M.; Levin, H.; McKee, A.; Ribbers, G.M.; Yaffe, K.; Zetterberg, H. Traumatic brain injuries. Nat. Rev. Dis. Primers 2016, 2, 16084. [Google Scholar] [CrossRef]
  231. Clinton, J.; Ambler, M.W.; Roberts, G.W. Post-traumatic alzheimer’s disease: Preponderance of a single plaque type. Neuropathol. Appl. Neurobiol. 1991, 17, 69–74. [Google Scholar] [CrossRef] [PubMed]
  232. Mortimer, J.A.; van Duijn, C.M.; Chandra, V.; Fratiglioni, L.; Graves, A.B.; Heyman, A.; Jorm, A.F.; Kokmen, E.; Kondo, K.; Rocca, W.A. Head trauma as a risk factor for alzheimer’s disease: A collaborative re-analysis of case-control studies. Eurodem risk factors research group. Int. J. Epidemiol. 1991, 20 (Suppl. 2), S28–S35. [Google Scholar] [CrossRef] [Green Version]
  233. Shahim, P.; Blennow, K.; Zetterberg, H. Tau, s-100 calcium-binding protein b, and neuron-specific enolase as biomarkers of concussion-reply. JAMA Neurol. 2014, 71, 926–927. [Google Scholar] [CrossRef] [PubMed]
  234. Shahim, P.; Tegner, Y.; Wilson, D.H.; Randall, J.; Skillback, T.; Pazooki, D.; Kallberg, B.; Blennow, K.; Zetterberg, H. Blood biomarkers for brain injury in concussed professional ice hockey players. JAMA Neurol. 2014, 71, 684–692. [Google Scholar] [CrossRef] [PubMed]
  235. Rubenstein, R.; Chang, B.; Davies, P.; Wagner, A.K.; Robertson, C.S.; Wang, K.K. A novel, ultrasensitive assay for tau: Potential for assessing traumatic brain injury in tissues and biofluids. J. Neurotrauma 2015, 32, 342–352. [Google Scholar] [CrossRef] [Green Version]
  236. Hu, W.T.; Watts, K.; Grossman, M.; Glass, J.; Lah, J.J.; Hales, C.; Shelnutt, M.; Van Deerlin, V.; Trojanowski, J.Q.; Levey, A.I. Reduced csf p-tau181 to tau ratio is a biomarker for ftld-tdp. Neurology 2013, 81, 1945–1952. [Google Scholar] [CrossRef] [Green Version]
  237. Vanmechelen, E.; Vanderstichele, H.; Davidsson, P.; Van Kerschaver, E.; Van Der Perre, B.; Sjögren, M.; Andreasen, N.; Blennow, K. Quantification of tau phosphorylated at threonine 181 in human cerebrospinal fluid: A sandwich elisa with a synthetic phosphopeptide for standardization. Neurosci. Lett. 2000, 285, 49–52. [Google Scholar] [CrossRef]
  238. Wagshal, D.; Sankaranarayanan, S.; Guss, V.; Hall, T.; Berisha, F.; Lobach, I.; Karydas, A.; Voltarelli, L.; Scherling, C.; Heuer, H.; et al. Divergent csf τ alterations in two common tauopathies: Alzheimer’s disease and progressive supranuclear palsy. J. Neurol. Neurosurg. Psychiatry 2015, 86, 244–250. [Google Scholar] [CrossRef]
  239. O’Brien, J.T.; Herholz, K. Amyloid imaging for dementia in clinical practice. BMC Med. 2015, 13, 163. [Google Scholar] [CrossRef]
  240. Henriksen, K.; O’Bryant, S.E.; Hampel, H.; Trojanowski, J.Q.; Montine, T.J.; Jeromin, A.; Blennow, K.; Lonneborg, A.; Wyss-Coray, T.; Soares, H.; et al. The future of blood-based biomarkers for alzheimer’s disease. Alzheimer’s Dement. J. Assoc. 2014, 10, 115–131. [Google Scholar] [CrossRef] [Green Version]
  241. de Almeida, S.M.; Shumaker, S.D.; LeBlanc, S.K.; Delaney, P.; Marquie-Beck, J.; Ueland, S.; Alexander, T.; Ellis, R.J. Incidence of post-dural puncture headache in research volunteers. Headache 2011, 51, 1503–1510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Mukaetova-Ladinska, E.B.; Abdell-All, Z.; Andrade, J.; da Silva, J.A.; Boksha, I.; Burbaeva, G.; Kalaria, R.N.; TO’Brien, J. Platelet tau protein as a potential peripheral biomarker in alzheimer’s disease: An explorative study. Curr. Alzheimer Res. 2018, 15, 800–808. [Google Scholar] [CrossRef] [PubMed]
  243. Slachevsky, A.; Guzmán-Martínez, L.; Delgado, C.; Reyes, P.; Farías, G.A.; Muñoz-Neira, C.; Bravo, E.; Farías, M.; Flores, P.; Garrido, C.; et al. Tau platelets correlate with regional brain atrophy in patients with alzheimer’s disease. J. Alzheimer’s Dis. 2017, 55, 1595–1603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Tzen, K.Y.; Yang, S.Y.; Chen, T.F.; Cheng, T.W.; Horng, H.E.; Wen, H.P.; Huang, Y.Y.; Shiue, C.Y.; Chiu, M.J. Plasma aβ but not tau is related to brain pib retention in early alzheimer’s disease. ACS Chem. Neurosci. 2014, 5, 830–836. [Google Scholar] [CrossRef]
  245. Zetterberg, H.; Blennow, K. Blood biomarkers: Democratizing alzheimer’s diagnostics. Neuron 2020, 106, 881–883. [Google Scholar] [CrossRef]
  246. Mattsson, N.; Zetterberg, H.; Janelidze, S.; Insel, P.S.; Andreasson, U.; Stomrud, E.; Palmqvist, S.; Baker, D.; Tan Hehir, C.A.; Jeromin, A.; et al. Plasma tau in alzheimer disease. Neurology 2016, 87, 1827–1835. [Google Scholar] [CrossRef] [Green Version]
  247. Mielke, M.M.; Hagen, C.E.; Wennberg, A.M.V.; Airey, D.C.; Savica, R.; Knopman, D.S.; Machulda, M.M.; Roberts, R.O.; Jack, C.R.; Petersen, R.C.; et al. Association of plasma total tau level with cognitive decline and risk of mild cognitive impairment or dementia in the mayo clinic study on aging. JAMA Neurol. 2017, 74, 1073–1080. [Google Scholar] [CrossRef]
  248. Dage, J.L.; Wennberg, A.M.V.; Airey, D.C.; Hagen, C.E.; Knopman, D.S.; Machulda, M.M.; Roberts, R.O.; Jack, C.R.; Petersen, R.C.; Mielke, M.M. Levels of tau protein in plasma are associated with neurodegeneration and cognitive function in a population-based elderly cohort. Alzheimer’s Dement. 2016, 12, 1226–1234. [Google Scholar] [CrossRef] [Green Version]
  249. Pereira, J.B.; Westman, E.; Hansson, O.; Initiative, A.s.D.N. Association between cerebrospinal fluid and plasma neurodegeneration biomarkers with brain atrophy in alzheimer’s disease. Neurobiol. Aging 2017, 58, 14–29. [Google Scholar] [CrossRef]
  250. Mielke, M.M.; Hagen, C.E.; Xu, J.; Chai, X.; Vemuri, P.; Lowe, V.J.; Airey, D.C.; Knopman, D.S.; Roberts, R.O.; Machulda, M.M.; et al. Plasma phospho-tau181 increases with alzheimer’s disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimer’s Dement. 2018, 14, 989–997. [Google Scholar] [CrossRef]
  251. Tatebe, H.; Kasai, T.; Ohmichi, T.; Kishi, Y.; Kakeya, T.; Waragai, M.; Kondo, M.; Allsop, D.; Tokuda, T. Quantification of plasma phosphorylated tau to use as a biomarker for brain alzheimer pathology: Pilot case-control studies including patients with alzheimer’s disease and down syndrome. Mol. Neurodegener. 2017, 12, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Yang, C.C.; Chiu, M.J.; Chen, T.F.; Chang, H.L.; Liu, B.H.; Yang, S.Y. Assay of plasma phosphorylated tau protein (threonine 181) and total tau protein in early-stage alzheimer’s disease. J. Alzheimer’s Dis. 2018, 61, 1323–1332. [Google Scholar] [CrossRef] [PubMed]
  253. Karikari, T.K.; Pascoal, T.A.; Ashton, N.J.; Janelidze, S.; Benedet, A.L.; Rodriguez, J.L.; Chamoun, M.; Savard, M.; Kang, M.S.; Therriault, J.; et al. Blood phosphorylated tau 181 as a biomarker for alzheimer’s disease: A diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol. 2020, 19, 422–433. [Google Scholar] [CrossRef]
  254. Thijssen, E.H.; La Joie, R.; Wolf, A.; Strom, A.; Wang, P.; Iaccarino, L.; Bourakova, V.; Cobigo, Y.; Heuer, H.; Spina, S.; et al. Diagnostic value of plasma phosphorylated tau181 in alzheimer’s disease and frontotemporal lobar degeneration. Nat. Med. 2020, 26, 387–397. [Google Scholar] [CrossRef] [PubMed]
  255. Janelidze, S.; Mattsson, N.; Palmqvist, S.; Smith, R.; Beach, T.G.; Serrano, G.E.; Chai, X.; Proctor, N.K.; Eichenlaub, U.; Zetterberg, H.; et al. Plasma p-tau181 in alzheimer’s disease: Relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s Dementia. Nat. Med. 2020, 26, 379–386. [Google Scholar] [CrossRef] [PubMed]
  256. Barthélemy, N.R.; Horie, K.; Sato, C.; Bateman, R.J. Blood plasma phosphorylated-tau isoforms track cns change in alzheimer’s disease. J. Exp. Med. 2020, 217, e20200861. [Google Scholar] [CrossRef]
  257. Lifke, V.; Kollmorgen, G.; Manuilova, E.; Oelschlaegel, T.; Hillringhaus, L.; Widmann, M.; von Arnim, C.A.F.; Otto, M.; Christenson, R.H.; Powers, J.L.; et al. Elecsys. Clin. Biochem. 2019, 72, 30–38. [Google Scholar] [CrossRef]
  258. Chiasserini, D.; Biscetti, L.; Farotti, L.; Eusebi, P.; Salvadori, N.; Lisetti, V.; Baschieri, F.; Chipi, E.; Frattini, G.; Stoops, E.; et al. Performance evaluation of an automated elisa system for alzheimer’s disease detection in clinical routine. J. Alzheimer’s Dis. 2016, 54, 55–67. [Google Scholar] [CrossRef]
  259. Rissin, D.M.; Fournier, D.R.; Piech, T.; Kan, C.W.; Campbell, T.G.; Song, L.; Chang, L.; Rivnak, A.J.; Patel, P.P.; Provuncher, G.K.; et al. Simultaneous detection of single molecules and singulated ensembles of molecules enables immunoassays with broad dynamic range. Anal Chem. 2011, 83, 2279–2285. [Google Scholar] [CrossRef] [Green Version]
  260. Barthélemy, N.R.; Fenaille, F.; Hirtz, C.; Sergeant, N.; Schraen-Maschke, S.; Vialaret, J.; Buée, L.; Gabelle, A.; Junot, C.; Lehmann, S.; et al. Tau protein quantification in human cerebrospinal fluid by targeted mass spectrometry at high sequence coverage provides insights into its primary structure heterogeneity. J. Proteome Res. 2016, 15, 667–676. [Google Scholar] [CrossRef]
  261. Barthélemy, N.R.; Mallipeddi, N.; Moiseyev, P.; Sato, C.; Bateman, R.J. Tau phosphorylation rates measured by mass spectrometry differ in the intracellular brain vs. Extracellular cerebrospinal fluid compartments and are differentially affected by alzheimer’s disease. Front. Aging Neurosci. 2019, 11, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Khalil, M.; Teunissen, C.E.; Otto, M.; Piehl, F.; Sormani, M.P.; Gattringer, T.; Barro, C.; Kappos, L.; Comabella, M.; Fazekas, F.; et al. Neurofilaments as biomarkers in neurological disorders. Nat. Rev. Neurol. 2018, 14, 577–589. [Google Scholar] [CrossRef] [PubMed]
  263. Gaetani, L.; Blennow, K.; Calabresi, P.; Di Filippo, M.; Parnetti, L.; Zetterberg, H. Neurofilament light chain as a biomarker in neurological disorders. J. Neurol. Neurosurg. Psychiatry 2019, 90, 870–881. [Google Scholar] [CrossRef] [PubMed]
  264. Bridel, C.; van Wieringen, W.N.; Zetterberg, H.; Tijms, B.M.; Teunissen, C.E.; Alvarez-Cermeño, J.C.; Andreasson, U.; Axelsson, M.; Bäckström, D.C.; Bartos, A.; et al. Diagnostic value of cerebrospinal fluid neurofilament light protein in neurology: A systematic review and meta-analysis. JAMA Neurol. 2019, 76, 1035–1048. [Google Scholar] [CrossRef] [PubMed]
  265. Weston, P.S.J.; Poole, T.; O’Connor, A.; Heslegrave, A.; Ryan, N.S.; Liang, Y.; Druyeh, R.; Mead, S.; Blennow, K.; Schott, J.M.; et al. Longitudinal measurement of serum neurofilament light in presymptomatic familial alzheimer’s disease. Alzheimer’s Res. Ther. 2019, 11, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Schultz, S.A.; Strain, J.F.; Adedokun, A.; Wang, Q.; Preische, O.; Kuhle, J.; Flores, S.; Keefe, S.; Dincer, A.; Ances, B.M.; et al. Serum neurofilament light chain levels are associated with white matter integrity in autosomal dominant alzheimer’s disease. Neurobiol. Dis. 2020, 142, 104960. [Google Scholar] [CrossRef]
  267. Kuhle, J.; Barro, C.; Andreasson, U.; Derfuss, T.; Lindberg, R.; Sandelius, Å.; Liman, V.; Norgren, N.; Blennow, K.; Zetterberg, H. Comparison of three analytical platforms for quantification of the neurofilament light chain in blood samples: Elisa, electrochemiluminescence immunoassay and simoa. Clin. Chem. Lab Med. 2016, 54, 1655–1661. [Google Scholar] [CrossRef]
  268. Kvartsberg, H.; Duits, F.H.; 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. Alzheimer’s Dement. 2015, 11, 1180–1190. [Google Scholar] [CrossRef]
  269. De Vos, A.; Jacobs, D.; Struyfs, H.; Fransen, E.; Andersson, K.; Portelius, E.; Andreasson, U.; De Surgeloose, D.; Hernalsteen, D.; Sleegers, K.; et al. C-terminal neurogranin is increased in cerebrospinal fluid but unchanged in plasma in alzheimer’s disease. Alzheimer’s Dement. 2015, 11, 1461–1469. [Google Scholar] [CrossRef] [Green Version]
  270. Ashton, N.J.; Nevado-Holgado, A.J.; Barber, I.S.; Lynham, S.; Gupta, V.; Chatterjee, P.; Goozee, K.; Hone, E.; Pedrini, S.; Blennow, K.; et al. A plasma protein classifier for predicting amyloid burden for preclinical alzheimer’s disease. Sci. Adv. 2019, 5, eaau7220. [Google Scholar] [CrossRef] [Green Version]
  271. Bouchon, A.; Dietrich, J.; Colonna, M. Cutting edge: Inflammatory responses can be triggered by trem-1, a novel receptor expressed on neutrophils and monocytes. J. Immunol. 2000, 164, 4991–4995. [Google Scholar] [CrossRef] [PubMed]
  272. Ulland, T.K.; Song, W.M.; Huang, S.C.; Ulrich, J.D.; Sergushichev, A.; Beatty, W.L.; Loboda, A.A.; Zhou, Y.; Cairns, N.J.; Kambal, A.; et al. Trem2 maintains microglial metabolic fitness in alzheimer’s disease. Cell 2017, 170, 649–663. [Google Scholar] [CrossRef] [PubMed]
  273. Schlepckow, K.; Kleinberger, G.; Fukumori, A.; Feederle, R.; Lichtenthaler, S.F.; Steiner, H.; Haass, C. An alzheimer-associated trem2 variant occurs at the adam cleavage site and affects shedding and phagocytic function. EMBO Mol. Med. 2017, 9, 1356–1365. [Google Scholar] [CrossRef]
  274. Yeh, F.L.; Wang, Y.; Tom, I.; Gonzalez, L.C.; Sheng, M. Trem2 binds to apolipoproteins, including apoe and clu/apoj, and thereby facilitates uptake of amyloid-beta by microglia. Neuron 2016, 91, 328–340. [Google Scholar] [CrossRef] [PubMed]
  275. Filipello, F.; Morini, R.; Corradini, I.; Zerbi, V.; Canzi, A.; Michalski, B.; Erreni, M.; Markicevic, M.; Starvaggi-Cucuzza, C.; Otero, K.; et al. The microglial innate immune receptor trem2 is required for synapse elimination and normal brain connectivity. Immunity 2018, 48, 979–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Kleinberger, G.; Yamanishi, Y.; Suárez-Calvet, M.; Czirr, E.; Lohmann, E.; Cuyvers, E.; Struyfs, H.; Pettkus, N.; Wenninger-Weinzierl, A.; Mazaheri, F.; et al. Trem2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci. Transl. Med. 2014, 6, 243ra286. [Google Scholar] [CrossRef]
  277. Guerreiro, R.; Wojtas, A.; Bras, J.; Carrasquillo, M.; Rogaeva, E.; Majounie, E.; Cruchaga, C.; Sassi, C.; Kauwe, J.S.; Younkin, S.; et al. Trem2 variants in alzheimer’s disease. N. Engl. J. Med. 2013, 368, 117–127. [Google Scholar] [CrossRef] [Green Version]
  278. Yaghmoor, F.; Noorsaeed, A.; Alsaggaf, S.; Aljohani, W.; Scholtzova, H.; Boutajangout, A.; Wisniewski, T. The role of trem2 in alzheimer’s disease and other neurological disorders. J. Alzheimer’s Dis. Parkinsonism 2014, 4, 160. [Google Scholar] [CrossRef]
  279. Jonsson, T.; Stefansson, H.; Steinberg, S.; Jonsdottir, I.; Jonsson, P.V.; Snaedal, J.; Bjornsson, S.; Huttenlocher, J.; Levey, A.I.; Lah, J.J.; et al. Variant of trem2 associated with the risk of alzheimer’s disease. N. Engl. J. Med. 2013, 368, 107–116. [Google Scholar] [CrossRef] [Green Version]
  280. Lill, C.M.; Rengmark, A.; Pihlstrøm, L.; Fogh, I.; Shatunov, A.; Sleiman, P.M.; Wang, L.S.; Liu, T.; Lassen, C.F.; Meissner, E.; et al. The role of trem2 r47h as a risk factor for alzheimer’s disease, frontotemporal lobar degeneration, amyotrophic lateral sclerosis, and parkinson’s disease. Alzheimer’s Dement. 2015, 11, 1407–1416. [Google Scholar] [CrossRef] [Green Version]
  281. Bekris, L.M.; Khrestian, M.; Dyne, E.; Shao, Y.; Pillai, J.A.; Rao, S.M.; Bemiller, S.M.; Lamb, B.; Fernandez, H.H.; Leverenz, J.B. Soluble trem2 and biomarkers of central and peripheral inflammation in neurodegenerative disease. J. Neuroimmunol. 2018, 319, 19–27. [Google Scholar] [CrossRef] [PubMed]
  282. Ashton, N.J.; Suárez-Calvet, M.; Heslegrave, A.; Hye, A.; Razquin, C.; Pastor, P.; Sanchez-Valle, R.; Molinuevo, J.L.; Visser, P.J.; Blennow, K.; et al. Plasma levels of soluble trem2 and neurofilament light chain in trem2 rare variant carriers. Alzheimer’s Res. Ther. 2019, 11, 94. [Google Scholar] [CrossRef] [PubMed]
  283. Piccio, L.; Deming, Y.; Del-Águila, J.L.; Ghezzi, L.; Holtzman, D.M.; Fagan, A.M.; Fenoglio, C.; Galimberti, D.; Borroni, B.; Cruchaga, C. Cerebrospinal fluid soluble trem2 is higher in alzheimer disease and associated with mutation status. Acta Neuropathol. 2016, 131, 925–933. [Google Scholar] [CrossRef] [PubMed]
  284. Suárez-Calvet, M.; Kleinberger, G.; Araque Caballero, M.; Brendel, M.; Rominger, A.; Alcolea, D.; Fortea, J.; Lleó, A.; Blesa, R.; Gispert, J.D.; et al. Strem2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early-stage alzheimer’s disease and associate with neuronal injury markers. EMBO Mol. Med. 2016, 8, 466–476. [Google Scholar] [CrossRef]
  285. Hakala, B.E.; White, C.; Recklies, A.D. Human cartilage gp-39, a major secretory product of articular chondrocytes and synovial cells, is a mammalian member of a chitinase protein family. J. Biol. Chem. 1993, 268, 25803–25810. [Google Scholar]
  286. Colton, C.A.; Mott, R.T.; Sharpe, H.; Xu, Q.; Van Nostrand, W.E.; Vitek, M.P. Expression profiles for macrophage alternative activation genes in ad and in mouse models of ad. J. Neuroinflam. 2006, 3, 27. [Google Scholar] [CrossRef] [Green Version]
  287. Johansen, J.S. Studies on serum ykl-40 as a biomarker in diseases with inflammation, tissue remodelling, fibroses and cancer. Dan Med. Bull. 2006, 53, 172–209. [Google Scholar]
  288. Johansen, J.S.; Schultz, N.A.; Jensen, B.V. Plasma ykl-40: A potential new cancer biomarker? Future Oncol. 2009, 5, 1065–1082. [Google Scholar] [CrossRef]
  289. Craig-Schapiro, R.; Perrin, R.J.; Roe, C.M.; Xiong, C.; Carter, D.; Cairns, N.J.; Mintun, M.A.; Peskind, E.R.; Li, G.; Galasko, D.R.; et al. Ykl-40: A novel prognostic fluid biomarker for preclinical alzheimer’s disease. Biol. Psychiatry 2010, 68, 903–912. [Google Scholar] [CrossRef] [Green Version]
  290. Mattsson, N.; Tabatabaei, S.; Johansson, P.; Hansson, O.; Andreasson, U.; Månsson, J.E.; Johansson, J.O.; Olsson, B.; Wallin, A.; Svensson, J.; et al. Cerebrospinal fluid microglial markers in alzheimer’s disease: Elevated chitotriosidase activity but lack of diagnostic utility. Neuromol. Med. 2011, 13, 151–159. [Google Scholar] [CrossRef]
  291. Teunissen, C.E.; Elias, N.; Koel-Simmelink, M.J.; Durieux-Lu, S.; Malekzadeh, A.; Pham, T.V.; Piersma, S.R.; Beccari, T.; Meeter, L.H.; Dopper, E.G.; et al. Novel diagnostic cerebrospinal fluid biomarkers for pathologic subtypes of frontotemporal dementia identified by proteomics. Alzheimer’s Dement. 2016, 2, 86–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Choi, J.; Lee, H.W.; Suk, K. Plasma level of chitinase 3-like 1 protein increases in patients with early alzheimer’s disease. J. Neurol. 2011, 258, 2181–2185. [Google Scholar] [CrossRef] [PubMed]
  293. Shen, X.N.; Niu, L.D.; Wang, Y.J.; Cao, X.P.; Liu, Q.; Tan, L.; Zhang, C.; Yu, J.T. Inflammatory markers in alzheimer’s disease and mild cognitive impairment: A meta-analysis and systematic review of 170 studies. J. Neurol. Neurosurg. Psychiatry 2019, 90, 590–598. [Google Scholar] [CrossRef] [PubMed]
  294. Park, J.C.; Han, S.H.; Mook-Jung, I. Peripheral inflammatory biomarkers in alzheimer’s disease: A brief review. BMB Rep. 2020, 53, 10–19. [Google Scholar] [CrossRef] [PubMed]
  295. Zuena, A.R.; Casolini, P.; Lattanzi, R.; Maftei, D. Chemokines in alzheimer’s disease: New insights into prokineticins, chemokine-like proteins. Front. Pharmacol. 2019, 10, 622. [Google Scholar] [CrossRef] [Green Version]
  296. Colangelo, A.M.; Alberghina, L.; Papa, M. Astrogliosis as a therapeutic target for neurodegenerative diseases. Neurosci. Lett. 2014, 565, 59–64. [Google Scholar] [CrossRef] [Green Version]
  297. Liddelow, S.; Barres, B. Snapshot: Astrocytes in health and disease. Cell 2015, 162, 1170. [Google Scholar] [CrossRef]
  298. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
  299. Liddelow, S.A. Modern approaches to investigating non-neuronal aspects of alzheimer’s disease. FASEB J. 2019, 33, 1528–1535. [Google Scholar] [CrossRef]
  300. Milà-Alomà, M.; Salvadó, G.; Gispert, J.D.; Vilor-Tejedor, N.; Grau-Rivera, O.; Sala-Vila, A.; Sánchez-Benavides, G.; Arenaza-Urquijo, E.M.; Crous-Bou, M.; González-de-Echávarri, J.M.; et al. Amyloid beta, tau, synaptic, neurodegeneration, and glial biomarkers in the preclinical stage of the alzheimer’s continuum. Alzheimer’s Dement. 2020. [Google Scholar] [CrossRef]
  301. Elahi, F.M.; Casaletto, K.B.; La Joie, R.; Walters, S.M.; Harvey, D.; Wolf, A.; Edwards, L.; Rivera-Contreras, W.; Karydas, A.; Cobigo, Y.; et al. Plasma biomarkers of astrocytic and neuronal dysfunction in early- and late-onset alzheimer’s disease. Alzheimer’s Dement. 2020, 16, 681–695. [Google Scholar] [CrossRef] [PubMed]
  302. James, B.D.; Wilson, R.S.; Boyle, P.A.; Trojanowski, J.Q.; Bennett, D.A.; Schneider, J.A. Tdp-43 stage, mixed pathologies, and clinical alzheimer’s-type dementia. Brain 2016, 139, 2983–2993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Majumder, V.; Gregory, J.M.; Barria, M.A.; Green, A.; Pal, S. Tdp-43 as a potential biomarker for amyotrophic lateral sclerosis: A systematic review and meta-analysis. BMC Neurol. 2018, 18, 90. [Google Scholar] [CrossRef] [PubMed]
  304. Feneberg, E.; Gray, E.; Ansorge, O.; Talbot, K.; Turner, M.R. Towards a tdp-43-based biomarker for als and ftld. Mol. Neurobiol. 2018, 55, 7789–7801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Huang, W.; Zhou, Y.; Tu, L.; Ba, Z.; Huang, J.; Huang, N.; Luo, Y. Tdp-43: From alzheimer’s disease to limbic-predominant age-related tdp-43 encephalopathy. Front. Mol. Neurosci. 2020, 13, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Meade, R.M.; Fairlie, D.P.; Mason, J.M. Alpha-synuclein structure and parkinson’s disease—Lessons and emerging principles. Mol. Neurodegener. 2019, 14, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Paciotti, S.; Bellomo, G.; Gatticchi, L.; Parnetti, L. Are we ready for detecting α-synuclein prone to aggregation in patients? The case of “protein-misfolding cyclic amplification” and “real-time quaking-induced conversion” as diagnostic tools. Front. Neurol. 2018, 9, 415. [Google Scholar] [CrossRef] [Green Version]
  308. Groveman, B.R.; Orrù, C.D.; Hughson, A.G.; Raymond, L.D.; Zanusso, G.; Ghetti, B.; Campbell, K.J.; Safar, J.; Galasko, D.; Caughey, B. Rapid and ultra-sensitive quantitation of disease-associated α-synuclein seeds in brain and cerebrospinal fluid by αsyn rt-quic. Acta Neuropathol. Commun. 2018, 6, 7. [Google Scholar] [CrossRef]
  309. Schweighauser, M.; Shi, Y.; Tarutani, A.; Kametani, F.; Murzin, A.G.; Ghetti, B.; Matsubara, T.; Tomita, T.; Ando, T.; Hasegawa, K.; et al. Structures of α-synuclein filaments from multiple system atrophy. Nature 2020, 1–6. [Google Scholar] [CrossRef]
  310. Montagne, A.; Zhao, Z.; Zlokovic, B.V. Alzheimer’s disease: A matter of blood-brain barrier dysfunction? J. Exp. Med. 2017, 214, 3151–3169. [Google Scholar] [CrossRef]
  311. Bennett, R.E.; Robbins, A.B.; Hu, M.; Cao, X.; Betensky, R.A.; Clark, T.; Das, S.; Hyman, B.T. Tau induces blood vessel abnormalities and angiogenesis-related gene expression in p301l transgenic mice and human alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2018, 115, E1289–E1298. [Google Scholar] [CrossRef] [Green Version]
  312. Snyder, H.M.; Corriveau, R.A.; Craft, S.; Faber, J.E.; Greenberg, S.M.; Knopman, D.; Lamb, B.T.; Montine, T.J.; Nedergaard, M.; Schaffer, C.B.; et al. Vascular contributions to cognitive impairment and dementia including alzheimer’s disease. Alzheimer’s Dement. 2015, 11, 710–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  313. Gottesman, R.F.; Schneider, A.L.; Zhou, Y.; Coresh, J.; Green, E.; Gupta, N.; Knopman, D.S.; Mintz, A.; Rahmim, A.; Sharrett, A.R.; et al. Association between midlife vascular risk factors and estimated brain amyloid deposition. JAMA 2017, 317, 1443–1450. [Google Scholar] [CrossRef] [PubMed]
  314. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef] [PubMed]
  315. Montagne, A.; Barnes, S.R.; Sweeney, M.D.; Halliday, M.R.; Sagare, A.P.; Zhao, Z.; Toga, A.W.; Jacobs, R.E.; Liu, C.Y.; Amezcua, L.; et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015, 85, 296–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Verheggen, I.C.M.; de Jong, J.J.A.; van Boxtel, M.P.J.; Gronenschild, E.H.B.M.; Palm, W.M.; Postma, A.A.; Jansen, J.F.A.; Verhey, F.R.J.; Backes, W.H. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience 2020, 42, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
  317. Nation, D.A.; Sweeney, M.D.; Montagne, A.; Sagare, A.P.; D’Orazio, L.M.; Pachicano, M.; Sepehrband, F.; Nelson, A.R.; Buennagel, D.P.; Harrington, M.G.; et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat. Med. 2019, 25, 270–276. [Google Scholar] [CrossRef]
  318. Wang, H.; Golob, E.J.; Su, M.Y. Vascular volume and blood-brain barrier permeability measured by dynamic contrast enhanced mri in hippocampus and cerebellum of patients with mci and normal controls. J. Magn. Reson. Imaging 2006, 24, 695–700. [Google Scholar] [CrossRef]
  319. Starr, J.M.; Farrall, A.J.; Armitage, P.; McGurn, B.; Wardlaw, J. Blood-brain barrier permeability in alzheimer’s disease: A case-control mri study. Psychiatry Res. 2009, 171, 232–241. [Google Scholar] [CrossRef]
  320. Sagare, A.P.; Sweeney, M.D.; Makshanoff, J.; Zlokovic, B.V. Shedding of soluble platelet-derived growth factor receptor-β from human brain pericytes. Neurosci. Lett. 2015, 607, 97–101. [Google Scholar] [CrossRef] [Green Version]
  321. Olsson, A.; Vanderstichele, H.; Andreasen, N.; De Meyer, G.; Wallin, A.; Holmberg, B.; Rosengren, L.; Vanmechelen, E.; Blennow, K. Simultaneous measurement of beta-amyloid(1-42), total tau, and phosphorylated tau (thr181) in cerebrospinal fluid by the xmap technology. Clin. Chem. 2005, 51, 336–345. [Google Scholar] [CrossRef] [PubMed]
  322. Tate, J.; Ward, G. Interferences in immunoassay. Clin. Biochem. Rev. 2004, 25, 105–120. [Google Scholar] [PubMed]
  323. Spector, R.; Robert Snodgrass, S.; Johanson, C.E. A balanced view of the cerebrospinal fluid composition and functions: Focus on adult humans. Exp. Neurol. 2015, 273, 57–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  324. Maiolini, R.; Bagrel, A.; Chavance, C.; Krebs, B.; Herbeth, B.; Masseyeff, R. Study of an enzyme immunoassay kit for carcinoembryonic antigen. Clin. Chem. 1980, 26, 1718–1722. [Google Scholar] [CrossRef]
  325. Pimm, M.V.; Perkins, A.C.; Armitage, N.C.; Baldwin, R.W. The characteristics of blood-borne radiolabels and the effect of anti-mouse igg antibodies on localization of radiolabeled monoclonal antibody in cancer patients. J. Nucl. Med. Off. Publ. Soc. Nucl. Med. 1985, 26, 1011–1023. [Google Scholar]
  326. Primus, F.J.; Kelley, E.A.; Hansen, H.J.; Goldenberg, D.M. “Sandwich”-type immunoassay of carcinoembryonic antigen in patients receiving murine monoclonal antibodies for diagnosis and therapy. Clin. Chem. 1988, 34, 261–264. [Google Scholar] [CrossRef]
  327. Thompson, R.J.; Jackson, A.P.; Langlois, N. Circulating antibodies to mouse monoclonal immunoglobulins in normal subjects--incidence, species specificity, and effects on a two-site assay for creatine kinase-mb isoenzyme. Clin. Chem. 1986, 32, 476–481. [Google Scholar] [CrossRef]
  328. d’Abramo, C.; Acker, C.M.; Schachter, J.B.; Terracina, G.; Wang, X.; Forest, S.K.; Davies, P. Detecting tau in serum of transgenic animal models after tau immunotherapy treatment. Neurobiol. Aging 2016, 37, 58–65. [Google Scholar] [CrossRef] [Green Version]
  329. Kricka, L.J. Human anti-animal antibody interferences in immunological assays. Clin. Chem. 1999, 45, 942–956. [Google Scholar] [CrossRef]
  330. Selby, C. Interference in immunoassay. Ann Clin Biochem 1999, 36 Pt 6, 704–721. [Google Scholar] [CrossRef]
  331. Wilkins, J.M.; Trushina, E. Application of metabolomics in alzheimer’s disease. Front. Neurol. 2017, 8, 719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  332. Trushina, E.; Mielke, M.M. Recent advances in the application of metabolomics to alzheimer’s disease. Biochim. Biophys. Acta 2014, 1842, 1232–1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Tambini, M.D.; Yao, W.; D’Adamio, L. Facilitation of glutamate, but not gaba, release in familial alzheimer’s app mutant knock-in rats with increased β-cleavage of app. Aging Cell 2019, 18, e13033. [Google Scholar] [CrossRef] [Green Version]
  334. Tambini, M.D.; D’Adamio, L. Knock-in rats with homozygous. J. Biol. Chem. 2020, 295, 7442–7451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Tambini, M.D.; Norris, K.A.; D’Adamio, L. Opposite changes in app processing and human aβ levels in rats carrying either a protective or a pathogenic app mutation. Elife 2020, 9, e52612. [Google Scholar] [CrossRef]
  336. Saito, T.; Matsuba, Y.; Mihira, N.; Takano, J.; Nilsson, P.; Itohara, S.; Iwata, N.; Saido, T.C. Single app knock-in mouse models of alzheimer’s disease. Nat. Neurosci. 2014, 17, 661–663. [Google Scholar] [CrossRef]
  337. Saito, T.; Mihira, N.; Matsuba, Y.; Sasaguri, H.; Hashimoto, S.; Narasimhan, S.; Zhang, B.; Murayama, S.; Higuchi, M.; Lee, V.M.Y.; et al. Humanization of the entire murine. J. Biol. Chem. 2019, 294, 12754–12765. [Google Scholar] [CrossRef]
  338. Tambini, M.D.; D’Adamio, L. Trem2 splicing and expression are preserved in a human aβ-producing, rat knock-in model of trem2-r47h alzheimer’s risk variant. Sci. Rep. 2020, 10, 4122. [Google Scholar] [CrossRef] [Green Version]
  339. Niedzwiecki, M.M.; Walker, D.I.; Howell, J.C.; Watts, K.D.; Jones, D.P.; Miller, G.W.; Hu, W.T. High-resolution metabolomic profiling of alzheimer’s disease in plasma. Ann. Clin. Transl. Neurol. 2020, 7, 36–45. [Google Scholar] [CrossRef] [Green Version]
  340. Ibáñez, C.; Simó, C.; Barupal, D.K.; Fiehn, O.; Kivipelto, M.; Cedazo-Mínguez, A.; Cifuentes, A. A new metabolomic workflow for early detection of alzheimer’s disease. J. Chromatogr. A 2013, 1302, 65–71. [Google Scholar] [CrossRef]
  341. Pomara, N.; Singh, R.; Deptula, D.; Chou, J.C.; Schwartz, M.B.; LeWitt, P.A. Glutamate and other csf amino acids in alzheimer’s disease. Am. J. Psychiatry 1992, 149, 251–254. [Google Scholar] [PubMed]
  342. Madeira, C.; Vargas-Lopes, C.; Brandão, C.O.; Reis, T.; Laks, J.; Panizzutti, R.; Ferreira, S.T. Elevated glutamate and glutamine levels in the cerebrospinal fluid of patients with probable alzheimer’s disease and depression. Front. Psychiatry 2018, 9, 561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Orešič, M.; Hyötyläinen, T.; Herukka, S.K.; Sysi-Aho, M.; Mattila, I.; Seppänan-Laakso, T.; Julkunen, V.; Gopalacharyulu, P.V.; Hallikainen, M.; Koikkalainen, J.; et al. Metabolome in progression to alzheimer’s disease. Transl. Psychiatry 2011, 1, e57. [Google Scholar] [CrossRef] [PubMed]
  344. Trushina, E.; Dutta, T.; Persson, X.M.; Mielke, M.M.; Petersen, R.C. Identification of altered metabolic pathways in plasma and csf in mild cognitive impairment and alzheimer’s disease using metabolomics. PLoS ONE 2013, 8, e63644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Kaddurah-Daouk, R.; Rozen, S.; Matson, W.; Han, X.; Hulette, C.M.; Burke, J.R.; Doraiswamy, P.M.; Welsh-Bohmer, K.A. Metabolomic changes in autopsy-confirmed alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 309–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  346. Mapstone, M.; Cheema, A.K.; Fiandaca, M.S.; Zhong, X.; Mhyre, T.R.; MacArthur, L.H.; Hall, W.J.; Fisher, S.G.; Peterson, D.R.; Haley, J.M.; et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nat. Med. 2014, 20, 415–418. [Google Scholar] [CrossRef]
  347. Frölich, L.; Peters, O.; Lewczuk, P.; Gruber, O.; Teipel, S.J.; Gertz, H.J.; Jahn, H.; Jessen, F.; Kurz, A.; Luckhaus, C.; et al. Incremental value of biomarker combinations to predict progression of mild cognitive impairment to Alzheimer’s Dementia. Alzheimer’s Res. Ther. 2017, 9, 84. [Google Scholar] [CrossRef] [Green Version]
  348. Spies, P.E.; Claassen, J.A.; Peer, P.G.; Blankenstein, M.A.; Teunissen, C.E.; Scheltens, P.; van der Flier, W.M.; Olde Rikkert, M.G.; Verbeek, M.M. A prediction model to calculate probability of alzheimer’s disease using cerebrospinal fluid biomarkers. Alzheimer’s Dement. 2013, 9, 262–268. [Google Scholar] [CrossRef]
  349. Antonaros, F.; Ghini, V.; Pulina, F.; Ramacieri, G.; Cicchini, E.; Mannini, E.; Martelli, A.; Feliciello, A.; Lanfranchi, S.; Onnivello, S.; et al. Plasma metabolome and cognitive skills in down syndrome. Sci. Rep. 2020, 10, 10491. [Google Scholar] [CrossRef]
  350. Orozco, J.S.; Hertz-Picciotto, I.; Abbeduto, L.; Slupsky, C.M. Metabolomics analysis of children with autism, idiopathic-developmental delays, and down syndrome. Transl. Psychiatry 2019, 9, 243. [Google Scholar] [CrossRef] [Green Version]
Table 1. Immunoassays and ultrasensitive platforms to detect tau in biological fluids.
Table 1. Immunoassays and ultrasensitive platforms to detect tau in biological fluids.
Type of AssayPlatform/
Manufacturer
t-taup-tauFluid
Sample
LOD
pg/mL
References
(PMID/link)
Standard ELISAINNOTEST
Fujirebio
Total tau
Antibodies:
Capture HT7/AT120
Detector BT2
CSF348748926
INNOTEST
Fujirebio
p-T181
Antibodies:
Capture HT7
Detector AT270
CSF1310788705
Applied Neurosolutions
(Dr. Peter Davies)
p-T231
Antibodies:
Capture CP27
Detector CP9
CSF926444757
Automated ELISAElecsysTotal tau
Antibodies:
Not available
CSF6331129184
Elecsys p-T181
Antibodies:
N.A.
CSF431129184
EUROIMMUNTotal tau
Antibodies:
Capture ADx201
Detector ADx215
CSF44.227447425
EUROIMMUN p-T181
Antibodies:
N.A.
CSFN.A.27447425
MSD-ECLMSD p-T181
Antibodies:
Capture biotinylated AT270
Detector SULFO-TAG-LRL (anti-tau, Lilly)
PlasmaN.A.29626426
MSD/Lilly p-T181
Antibodies:
Capture biotinylated AT270
Detector SULFO-TAG-LRL (anti-tau)
CSFN.A.31709776
MSD/Lilly p-T217
Antibodies:
Capture biotinylated IBA413
Detector SULFO-TAG-LRL (anti-tau)
CSFN.A.31709776
MSD/Lilly p-T217
Antibodies:
Capture Biotinylated-IBA493
Detector SULFO-TAG-4G10-E2 (anti-tau)
PlasmaN.A.31709776
SiMoASimoa™ Tau 2.0 kit
HD-1 platform
(Quanterix)
Total tau
Antibodies:
Capture sequence AA16–AA24
Detector sequence AA218–AA222
CSF0.019101444
Janssen R&D
Simoa technology
HD-1 platform
(Quanterix)
Total tau
Antibodies:
Capture HT7
Detector PT82
CSFN.A.32246036
Modiefied version of Simoa™ Tau 2.0 kit
HD-1 platform
(Quanterix)
p-T181
Antibodies:
Capture sequence AA16–AA24
Detector AT270
Plasma0.009028866979
Sandwich immunoassay format on Simoa technology
(Quanterix)
p-T181
Antibodies:
Capture AT270
Detector Tau12
Plasma
Serum
N.A.32333900
Simoa™ p-181 kit
HD-1 platform
(Quanterix)
NEW
p-T181
Antibodies: N/A
CSDF
Plasma
Serum
Alzheimer Association International Conference, 2020. Available online: https://alz.confex.com/alz/20amsterdam/meetingapp.cgi/Paper/41238. Accessed on 29 July 2020.
Janssen R&D
Simoa technology
(Quanterix)
p-T217
Antibodies:
Capture PT3
Detector PT82
CSFN.A.32246036
Simoa® pTau-231
Advantage Kit
HD-1 platform
(Quanterix)
p-T231
Antibodies:
Capture AT270
Detector Tau12
CSF0.621Quanterix.com. Available online: https://www.quanterix.com/sites/default/files/assays/Simoa_pTau-231_Data_Sheet_HD-1_HD-X_Rev02.pdf. Accessed on 29 July 2020.
NT1 assay
Simoa technology
HD-1 platform
(Quanterix)
N-terminal tau
Antibodies:
Capture Tau12
Detector BT2
CSF
Plasma
0.2-0.730419228
Nano liquid chromatography-HRMS Total taup-T181
p-S202
p-T205
p-T217
CSFN.A.26742856
32161412
(IP)-LC-MS p-T217
p-T181
Plasmap-T217:0.05
p-T181:0.2
32725127
SQUID-IMR Total taup-T181PlasmaN.A.29376870
A detailed list of the original and the more updated assays used in the tau biomarker field is provided, with emphasis on total, phosphorylated or truncated tau. Type of assays and platforms: ELISA (Enzyme-linked immunosorbent assay); Meso Scale Discovery (MSD) technology with electrochemiluminescence; SiMoA: Single-Molecule enzyme-linked immunosorbent assay; Nano liquid chromatography–HRMMSD-ECLS: Nano liquid chromatography–high resolution mass spectrometry; (IP)-LC-MS: immunoprecipitation coupled with liquid chromatography-spectrometry; SQUID-IMR: superconducting-quantum-interference-device Immunomagnetic reduction. Fluid samples are specified (CSF, plasma, serum). Limit of detection (LOD) of the assay is provided where available, if not it is marked as N.A. References are listed as PMID or hyperlink.
Table 2. Multi-target platforms.
Table 2. Multi-target platforms.
Type of AssayPlatform/
Manufacturer
TargetsFluid
Sample
LOD
pg/mL
References
(PMID/link)
SiMoASimoa® Neurology 3-Plex A Kit
HD-1 platform
(Quanterix)
t-tau
Aβ40
Aβ42
CSF
plasma
Tau: 0.019
Aβ40: 0.196
Aβ42: 0.045
Quanterix.com. Available online: https://www.quanterix.com/sites/default/files/assays/Simoa_N3PA_Data_Sheet_HD-1_HD-X_Rev04%20%281%29.pdf. Accessed on 29 July 2020
Simoa® Neurology 4-Plex A Kit
HD-1 platform
(Quanterix)
t-tau
NfL
GFAP
UCLH-1
CSF
plasma
Tau: 0.024
NfL: 0.104
GFAP: 0.221
UCLH-1: 1.74
Quanterix.com. Available online: https://www.quanterix.com/sites/default/files/assays/Simoa_N4PA_Data_Sheet_HD-1_HD-X_DS-0074_rev7.pdf. Accessed on 29 July 2020
Simoa® 4X-plex neurology Kit
(Quanterix)
NEW
Aβ40
Aβ42
GFAP
NfL
CSF
plasma
Aβ40: 0.084
Aβ42: 0.148
GFAP: 1.66
NfL: 1.157
Alzheimer Association International Conference, 2020. Available online: https://alz.confex.com/alz/20amsterdam/meetingapp.cgi/Paper/43506. Accessed on 29 July 2020.
Luminex®xMAP®The MILLIPLEX® map
4X multiplex immunoassay kit
(Millipore, Sigma)
t-tau
p-T181
Aβ40
Aβ42
CSFN.A.Emdmillipore.com. Available online: https://www.emdmillipore.com/US/en/product/MILLIPLEX-MAP-Human-Amyloid-Beta-and-Tau-Magnetic-Bead-Panel-Multiplex-Assay,MM_NF-HNABTMAG-68K. Accessed on 29 July 2020.
NeuroToolKitRoche’s Elecsys electroluminescence immunoassay platformt-tau, p-T181
Aβ40, Aβ42
α-synuclein
S100b
YKL-40
GFAP
sTREM2
IL-6
NfL
Neurogranin
CSFN.A.32573951
Listed are some of the assays available for the simultaneous detection of multiple biomarkers in CSF or plasma. Type of assays and platforms: SiMoA: Single-Molecule enzyme-linked immunosorbent assay; Luminex XMAP: bead-based multiplex immunoassay in a microplate format; NeuroToolKit: based on electroluminescence immunoassay platform. Targets and Fluid samples are specified. Limit of detection (LOD) of the assay is provided when available. References are listed as PMID or hyperlink.

Share and Cite

MDPI and ACS Style

d’Abramo, C.; D’Adamio, L.; Giliberto, L. Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer’s Disease: Sensitivity, Specificity and Potential for Clinical Use. J. Pers. Med. 2020, 10, 116. https://doi.org/10.3390/jpm10030116

AMA Style

d’Abramo C, D’Adamio L, Giliberto L. Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer’s Disease: Sensitivity, Specificity and Potential for Clinical Use. Journal of Personalized Medicine. 2020; 10(3):116. https://doi.org/10.3390/jpm10030116

Chicago/Turabian Style

d’Abramo, Cristina, Luciano D’Adamio, and Luca Giliberto. 2020. "Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer’s Disease: Sensitivity, Specificity and Potential for Clinical Use" Journal of Personalized Medicine 10, no. 3: 116. https://doi.org/10.3390/jpm10030116

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop