Skip to main content

REVIEW article

Front. Neurosci., 27 April 2022
Sec. Neurodegeneration
This article is part of the Research Topic Mechanisms of Action in Neurodegenerative Proteinopathies View all 8 articles

Alzheimer’s Disease Biomarkers Revisited From the Amyloid Cascade Hypothesis Standpoint

  • 1Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
  • 2UK Dementia Research Institute at UCL, London, United Kingdom
  • 3Department of Neurodegenerative Disease, Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
  • 4Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
  • 5Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden

Alzheimer’s disease (AD) is the most common neurodegenerative disease worldwide. Amyloid beta (Aβ) is one of the proteins which aggregate in AD, and its key role in the disease pathogenesis is highlighted in the amyloid cascade hypothesis, which states that the deposition of Aβ in the brain parenchyma is a crucial initiating step in the future development of AD. The sensitivity of instruments used to measure proteins in blood and cerebrospinal fluid has significantly improved, such that Aβ can now successfully be measured in plasma. However, due to the peripheral production of Aβ, there is significant overlap between diagnostic groups. The presence of pathological Aβ within the AD brain has several effects on the cells and surrounding tissue. Therefore, there is a possibility that using markers of tissue responses to Aβ may reveal more information about Aβ pathology and pathogenesis than looking at plasma Aβ alone. In this manuscript, using the amyloid cascade hypothesis as a starting point, we will delve into how the effect of Aβ on the surrounding tissue can be monitored using biomarkers. In particular, we will consider whether glial fibrillary acidic protein, triggering receptor expressed on myeloid cells 2, phosphorylated tau, and neurofilament light chain could be used to phenotype and quantify the tissue response against Aβ pathology in AD.

Introduction

Alzheimer’s disease (AD) is a neurodegenerative disease characterised by three key neuropathological hallmarks: (1) the presence of amyloid beta (Aβ) plaques in the brain parenchyma, and commonly also within cerebral blood vessels (cerebral amyloid angiopathy); (2) intraneuronal neurofibrillary tangles (NFTs), composed of hyperphosphorylated tau; and (3) neurodegeneration (Stelzmann et al., 1995; DeKosky, 2001; Winner et al., 2011). The accumulation of misfolded proteins, in the form of Aβ plaques and NFTs, has led to AD being termed a proteinopathy (Villemagne et al., 2018). However, of particular interest is the role Aβ plays in AD pathogenesis.

The amyloid cascade hypothesis, described by Hardy and Higgins (1992), introduced the idea of Aβ misfolding and deposition being the primary precipitant of AD, with the other neuropathological hallmarks, including NFT formation and neuronal loss, occurring as a direct consequence of the misfolded Aβ. Along with Aβ misfolding, Hardy and Selkoe (2002) also describe an imbalance between Aβ production and clearance, resulting in an increased presence of cerebral Aβ in its various forms, including monomers, oligomers, insoluble fibrils and plaques (Mawuenyega et al., 2010). Evidence in favour of this hypothesis can be seen in trisomy 21, more commonly known as Down syndrome (DS), as well as in the Icelandic Aβ precursor protein (APP) mutation. DS is caused by individuals having three copies of chromosome 21, the same chromosome that houses the APP gene. It is these additional copies of APP which contribute to DS being the leading genetic cause of AD (Wiseman et al., 2015). Conversely, the A673T point mutation in APP, frequently termed the Icelandic mutation, is the first APP mutation known to be protective against Aβ deposition and AD (Jonsson et al., 2012). This mutation reduces cleavage of APP by β-secretase along its amyloidogenic pathway, and produces Aβ that is less prone to aggregation (Maloney et al., 2014). However, given the multi-factorial nature of AD, it is important to acknowledge that controversies exist regarding the plausibility of the amyloid cascade hypothesis (Morris et al., 2014). Indeed, there is a possibility that the accumulation of by-products of Aβ production, such as the 99 amino acid C-terminal fragment of APP, or even the neuroprotective nature of shorter Aβ peptide fragments, may play a large role in AD pathogenesis (Moore et al., 2018; Pera et al., 2020). Furthermore, there is growing evidence in favour of alternative hypotheses which place other proteins or processes, such as tau and neuroinflammation, as the central initiating mechanisms of AD pathogenesis (Alvarez A. et al., 1999; Alvarez R. et al., 1999; Ittner and Götz, 2011; Guzman-Martinez et al., 2019).

It is now clear that AD neuropathology begins up to 20 years before symptom-onset (Reiman et al., 2012; Villemagne et al., 2013). Furthermore, cerebrospinal fluid (CSF) and plasma Aβ are the first fluid biomarkers to significantly change in AD patients, and they do so prior to Aβ positron emission tomography (PET) positivity (Palmqvist et al., 2019). Therefore, increasing our understanding of this protein is essential to enable us to identify treatments which target AD at its root. In this review, we will consider Aβ deposition as one of the causes of AD, looking at its various isoforms, methods for its detection in biofluids, and biomarkers of tissue reactions to Aβ that could be used as indirect measures of Aβ pathology, and to improve our understanding of Aβ toxicity.

Amyloid-β Formation, Aggregation and Isoforms

Aβ is a peptide that is naturally present within the healthy human brain, where it is produced intracellularly and at the cell membrane, and subsequently released into the extracellular space (Finder and Glockshuber, 2007). However, pathogenic Aβ is produced when APP, a transmembrane protein, is sequentially cleaved along its amyloidogenic pathway by β- and γ-secretases (Cacace et al., 2016). The site of γ-secretase cleavage determines the length of the resultant Aβ peptide (Perrone et al., 2020), ranging from 37 to 49 amino acids in length (Chen et al., 2017). The most abundant isoforms of Aβ in CSF are Aβ1–38, Aβ1–40, and Aβ1–42 (Struyfs et al., 2015), with the 40 and 42 amino acid length isoforms being the two most widely researched. This is predominantly due to the important role that the Aβ1–42/Aβ1–40 ratio (Aβ1–42/1–40) plays in supporting a diagnosis of AD, as highlighted by the amyloid, tau, neurodegeneration, or AT(N), diagnostic criteria for AD (Jack et al., 2016, 2018). Whilst CSF concentrations of Aβ1–40 remain unchanged in AD, Aβ1–42 concentrations decrease, which is thought to reflect aggregation and deposition within the brain (Lewczuk et al., 2004). Therefore, looking at the two in combination, as a ratio, provides a more accurate marker of plaque pathology in comparison to the overall Aβ production in that individual, and combats issues of inter-individual baseline concentration differences posed by looking at CSF Aβ1–42 concentrations in isolation (Alawode et al., 2021).

However, longer-length peptides, such as Aβ1–43, have been observed more frequently than Aβ1–40 within Aβ plaque cores in familial AD (FAD), sporadic AD and Down syndrome brains (Hirayama et al., 2003; Welander et al., 2009; Keller et al., 2010). Indeed, post-mortem analysis of AD brains has revealed a positive correlation between Aβ peptide length and plaque load (Aβ1–43 > Aβ1–42 > Aβ1–40) (Jäkel et al., 2019), and studies in mouse models of FAD have revealed that Aβ1–43 is more neurotoxic, and has a greater propensity to aggregate, than Aβ1–42 (Saito et al., 2011). Furthermore, CSF concentrations of Aβ1–43 are significantly reduced in FAD mutation carriers (Perrone et al., 2020), mimicking the reduction in Aβ1–42 seen in AD, and highlighting a potential role of Aβ1–43 in the disease.

As alluded to above, variability in the C-terminus of Aβ is a well-known phenomenon. However, there is similar heterogeneity at the N-terminus of the peptide (Colin et al., 1985; Miller et al., 1993). In fact, investigations have revealed that only a small proportion of the Aβ ending at amino acids 40 and 42 within cerebral blood vessels and parenchymal plaques, respectively, is made up by Aβ1–40 and Aβ1–42 (Harigaya et al., 2000), highlighting the overriding presence of truncated or modified Aβ in AD brains. One such truncated species, pyroglutamate-modified Aβ (AβpE), is one of the dominant forms of Aβ in the hippocampi and cortices of AD patients (Portelius et al., 2010), and compared to full-length Aβ, irrespective of the C-terminus, AβpE that has been truncated and modified at the third amino acid of Aβ (AβpE3) has shown an up to 250-fold increased rate of aggregation (Schilling et al., 2006). Additionally, AβpE is the only identified form of Aβ that is solely found within plaques, and is not produced by neurones (DeMattos et al., 2012), making it a plaque-specific form of Aβ and a promising immunotherapy target.

Monomeric Aβ exists in both an α-helical and β-pleated sheet conformation, and is amphiphatic in nature, exhibiting hydrophilicity at the N-terminal amino acids, and hydrophobicity at the C-terminus (Finder and Glockshuber, 2007). These monomeric isoforms can subsequently aggregate to form: (1) soluble oligomers, which are heterogenous in size and can spread throughout the brain; (2) protofibrils, which are larger soluble oligomers; or (3) insoluble fibrils, which can further aggregate to form Aβ plaques (Chen et al., 2017; Hampel et al., 2021). All of these aggregated forms of Aβ are known to be neurotoxic (Kuchibhotla et al., 2008; Koffie et al., 2009; Meyer-Luehmann et al., 2009; Hampel et al., 2021). Fibril formation is now widely considered to occur by nucleation-dependent polymerisation (Chatani and Yamamoto, 2018; Pyun et al., 2020). This process involves the initial formation of nuclei, followed by an elongation phase, resulting in fibril formation (Finder and Glockshuber, 2007; Chatani and Yamamoto, 2018). Furthermore, this process is concentration dependent. However, Aβ1–42 is much more prone to aggregation, requiring a five-fold lower minimum concentration to aggregate into fibrils than Aβ1–40, highlighting why Aβ1–42 is much more abundant in plaques than Aβ1–40 (Finder and Glockshuber, 2007).

Tissue Reaction to Amyloid-β Pathology

The presence and accumulation of pathogenic Aβ plaques in the brain parenchyma of AD patients has several neurotoxic effects on the surrounding tissue. Firstly, Aβ triggers an inflammatory response mediated by glial cells within the central nervous system (CNS) (Cai et al., 2014). There is increasing evidence showing that neuroinflammation, and the activation of a variety of CNS-specific glial cells, is emerging as a central player in AD pathogenesis and neuropathology (Heneka et al., 2015; Bronzuoli et al., 2016). As the primary immunosurveillance cells of the CNS, microglia are the first cells to be activated in response to foreign material within the brain. In contrast to microglia, astrocytes function predominantly as a neurosupportive cell type, contributing largely to synaptogenesis, and maintaining synapse and blood-brain barrier (BBB) integrity (Perea et al., 2009; Bronzuoli et al., 2016). When they become activated, microglia secrete pro-inflammatory cytokines, and act as the macrophages of the brain to clear the abnormal debris (Cameron and Landreth, 2009; Ransohoff and Perry, 2009). Furthermore, studies in mouse models of AD have revealed the presence of dystrophic neurites in close proximity to Aβ plaques, which both microglia and astrocytes respond to in an attempt to repair the damage (Sanchez-Varo et al., 2011). Indeed, in the presence of AD neuropathology, both microglia and astrocytes adopt a reactive phenotype, termed reactive gliosis, which has both neuroprotective and neurotoxic effects within AD brains (Bronzuoli et al., 2016). However, the uncontrolled and prolonged activation of both of these glial cells due to increasing Aβ burden eventually leads to their dysfunction, promotes chronic neuroinflammation, and contributes to the loss of synapses around plaques (Canevari et al., 2004; Subramanian et al., 2020).

Secondly, the presence of Aβ induces oxidative stress and disrupts calcium homeostasis, leading to neuronal toxicity (Canevari et al., 2004). This toxicity results in necrotic neuronal death, and the resultant decrease in brain volume associated with AD, particularly in the hippocampi (Behl et al., 1994). Along with the loss of synapses, this neurodegeneration significantly contributes to the cognitive decline seen in AD patients (Deture and Dickson, 2019).

Thirdly, Aβ induces the intraneuronal phosphorylation of tau (a protein that is microtubule-associated in its non-phosphorylated state), its subsequent aggregation to form NFTs, and its extracellular secretion (Busciglio et al., 1995; Mattsson-Carlgren et al., 2020). Tau phosphorylation, which may eventually cause NFT pathology, disrupts the neuronal cytoskeleton, and similar to Aβ, contributes to synapse loss and cognitive decline (Gómez-Isla et al., 1997; Metaxas and Kempf, 2016).

In essence, Aβ pathology, and the resultant tissue reactions to this peptide, are known processes that are now possible to monitor and measure in biofluids as well as with PET neuroimaging. This has been made possible due to the recent developments in supersensitive immunoassay and mass spectrometry (MS) detection methods.

Methods for Fluid Biomarker Measurement

Methods for fluid biomarker measurement have been around for many years. With respect to Aβ and its tissue reactions, they have largely involved the use of enzyme-linked immunosorbent assay (ELISA) (Sjögren et al., 2001; Welge et al., 2009; Ishiki et al., 2016; Piccio et al., 2016). However, as we move into measuring Aβ and markers of its tissue reactions in blood, there is a need to use more sensitive detection methods. Hence, for the purposes of this review, we will focus on Single molecule array (Simoa) technology and MS.

Single Molecule Array

Simoa is a form of digital ELISA that measures the fluorescence from single enzyme-labelled protein molecules conjugated onto magnetic beads, and trapped within femtolitre-sized wells (Rissin et al., 2010). Whilst conventional ELISA is currently the gold standard technique for detecting and quantifying proteins, digital ELISA using Simoa is emerging as an ultrasensitive method for protein detection, increasing sensitivity of protein detection from picomolar (10–12 M) to subfemtomolar (10–15 M) concentrations (Rissin et al., 2010; Cohen and Walt, 2019; Kan et al., 2020). This is particularly important as the serum concentration of proteins involved in AD are thought to range from 10–16 to 10–12 M (Galasko, 2005; Jong et al., 2007). Simoa measurements of Aβ, glial fibrillary acidic protein (GFAP), phosphorylated tau (p-tau) and neurofilament light chain (NfL), but not the soluble fragment of triggering receptor expressed on myeloid cells 2 (sTREM2), in both CSF and plasma are now well-established (Janelidze et al., 2016; Hendricks et al., 2019; Vergallo et al., 2019; Benedet et al., 2021; Blennow, 2021; Park et al., 2021). However, as we move into the era of disease-modifying therapies for AD targeting Aβ pathology, there is a need for more sensitive and robust techniques to measure changes in these proteins in trial participants. Furthermore, a more sensitive detection method provides the possibility of diluting samples prior to analysis, thereby reducing matrix effects without compromising detectability of proteins of interest (Song et al., 2015). One possible solution to this ongoing issue of sensitivity is the recent development of upgraded Simoa technology, which can measure proteins down to sub-attomolar (10–18 M) concentrations (Kan et al., 2020). This increased sensitivity has been achieved primarily by: (1) increasing the molecule:bead ratio through the use of fewer beads (5,000 compared to 500,000 in conventional Simoa); and (2) using magnetic-meniscus sweeping to increase the proportion of beads loaded into the microwells (Kan et al., 2020). Although this upgrade is not yet commercially available, preliminary investigations have revealed an increase in sensitivity of up to 100-fold provided by this technique compared to conventional Simoa (Kan et al., 2020).

Mass Spectrometry

MS is an analytical technique for measuring the mass-to-charge ratio of analyte ions, thus, by nature, it can achieve a greater specificity compared to immunoassays. Standard MS workflow involves an initial separation step of the analyte prior to analysis and detection within the mass spectrometer. Various systems can be coupled to the mass spectrometer in order to achieve this separation, including liquid chromatography and gas chromatography (Smith, 2013). One key difference compared to immunoassays is that the mass spectrometer is antibody-independent. This can be important where there are no suitable antibodies for detection purposes, although antibodies can be utilised to enrich samples by immunoprecipitation prior to the mass spectrometry step. Denaturing conditions, used in sample preparation, as well as the samples being handled in aqueous organic solvents, mean results obtained can be less influenced by matrix effects than in an immunoassay (Pannee et al., 2014; Crutchfield et al., 2016; Oeckl and Otto, 2019).

Arguably of most interest with regards to Aβ is a direct comparison of MS with Simoa-based quantification of Aβ1–40 and Aβ1–42 in a preclinical AD cohort, conducted by Keshavan et al. (2021), which observed that at this stage of disease, MS measurements showed a higher correlation with brain Aβ pathology than Simoa measurements. Further comparisons between MS and other immunoassay techniques have shown similar results. A head-to-head comparison of eight plasma Aβ1–42/1–40 assays, including four MS and four immunoassays, revealed that MS methods for plasma Aβ1–42/1–40 measurement provide greater discriminative accuracy between Aβ-positive and Aβ-negative individuals, as measured by Aβ PET (Janelidze et al., 2021). Furthermore, MS correlates better with CSF Aβ1–42/1–40 measurements than immunoassay methods in two independent disease cohorts (Janelidze et al., 2021). In particular, immunoprecipitation-MS developed by Randall Bateman at Washington University was observed as the most superior in all aspects assessed. The promising results observed with MS in comparison to immunoassays opens questions as to whether MS may be the future of Aβ measurements. Furthermore, MS methods for detection of p-tau, and sTREM2 have also shown promise (Heslegrave et al., 2016; Korecka and Shaw, 2021), with little to no published work on MS detection of GFAP or NfL. However, it is important to consider the space, cost and sample preparation time required for a mass spectrometer, and whether the differences between MS and immunoassays are significant enough to warrant a complete change to purely MS-based analyses of these proteins.

Apparent Fluid Biomarkers for Amyloid-β Pathology

Historically, AD diagnoses were made based on symptoms alone. However, studies have revealed that the neuropathological hallmarks of AD begin up to 20 years prior to symptom onset (Reiman et al., 2012; Villemagne et al., 2013). As such, there has been a move towards using a biological definition of disease to support a clinical diagnosis of AD (Jack et al., 2018). The AT(N) criteria for AD diagnosis draws together the three key pathophysiological characteristics of AD using both fluid and neuroimaging biomarkers, and is described by Jack et al. (2016). However, for the purposes of this review, we will focus solely on the fluid biomarkers of Aβ pathology. In particular, we will consider direct fluid biomarkers of Aβ pathology – Aβ1–42/1–40 and Aβ1–43 – and markers of tissue reactions to Aβ – GFAP, triggering receptor expressed on myeloid cells 2 (TREM2), p-tau, and NfL.

Direct Markers of Amyloid-β Pathology

Amyloid-β 1–40, 1–42, 1–42/1–40 and 1–43

As highlighted by Jack et al. (2016), the fluid biomarkers recognised by the AT(N) criteria for “A” or Aβ pathology are CSF Aβ1–42 or Aβ1–42/1–40. To place an individual on the AD continuum, they must have evidence of positive Aβ pathology, whether that be through a reduction in the CSF biomarkers or a positive Aβ PET scan. The absence of positive Aβ biomarkers is suggestive of a non-AD pathological change (Jack et al., 2018). The recent update of the AlzBiomarker database comprises a meta-analysis of studies comparing biomarker levels in AD vs controls, as well as cross-disease comparisons (Accessed 7th December 2021)1 (Olsson et al., 2016). The database reveals that in AD vs controls, CSF Aβ1–40 decreases by 9%, whereas CSF Aβ1–42 decreases by 45%, highlighting a much larger decrease in CSF Aβ1–42 in AD. Both of these percentage decreases are statistically significant.

Studies on CSF concentrations of Aβ1–43 have revealed this peptide, as well as CSF Aβ1–43/1–40, to be significantly reduced in both AD compared to controls and early-onset AD compared to late-onset AD (Lauridsen et al., 2017; Perrone et al., 2020). However, in contrast to Aβ1–40 and Aβ1–42, the quantity of studies investigating CSF Aβ1–43 remains relatively scarce. One possible reason for this is that some studies have observed little difference between the diagnostic accuracy of Aβ1–42 and Aβ1–43 (Bruggink et al., 2013; Lauridsen et al., 2017). Therefore, it could be argued that given the already established robustness of CSF Aβ1–42 and Aβ1–42/1–40 in AD diagnosis, there is no place for Aβ1–43. However, one study found CSF Aβ1–43, but not CSF Aβ1–42, could identify patients with amnestic MCI who later progressed to AD, as well as observing a significant decrease in CSF Aβ1–43 concentrations over the 2-year follow-up period, compared to no significant differences in CSF Aβ1–42 concentrations (Lauridsen et al., 2016). Furthermore, a separate study observed CSF Aβ1–43 concentrations to be significantly reduced to a greater extent in early onset AD compared to late onset AD – a finding not true to CSF Aβ1–42 (Lauridsen et al., 2017). Together, these studies highlight a potential role of Aβ1–43, albeit less well investigated, in distinguishing between AD diagnostic groups (Alawode et al., 2021).

Aβ PET has been used to assess the presence or absence of Aβ pathology for several years, and was used to validate the sensitivity and efficacy of the CSF Aβ biomarkers (Blennow et al., 2015). Whilst the ability to detect the presence of Aβ pathology using CSF has been invaluable, there is a need to develop blood-based biomarkers for diagnosis. This is because blood measurements incur much lower costs and are more easily accessible in low-resource and non-specialist settings worldwide (Molinuevo et al., 2018; Shi et al., 2018; Albani et al., 2019). Additionally, blood can be obtained less invasively than CSF, so could function as an initial diagnostic screening tool in the primary care setting, followed by more in-depth analysis in specialist centres (Janelidze et al., 2016; Molinuevo et al., 2018). However, drawbacks evidently exist in measuring biomarkers of brain diseases using peripheral body fluids, namely: (1) the BBB results in a 10–100 fold lower concentration of the analytes compared to in CSF (Blennow and Zetterberg, 2015); (2) some AD biomarkers are expressed by extra-cerebral tissues; (3) analytes of interest may undergo degradation by blood proteases prior to their measurement (Zetterberg and Burnham, 2019). Indeed, whilst Aβ is most commonly discussed with regards to AD, Aβ production appears to occur in all cells and tissues of the body (Li et al., 1999), and deposits have been observed in extra-cerebral tissues, including systemic blood vessels, platelets, skin, subcutaneous tissue, intestines and the eye (Mori et al., 1989; Li et al., 1999; Hart et al., 2016). Plasma and serum Aβ1–40, Aβ1–42, or Aβ1–42/1–40 measurements have been investigated as potential blood biomarkers for AD. However, despite plasma Aβ concentrations fluctuating with time, continuous contributions from extra-cerebral tissues mean that plasma Aβ concentrations do not change as dynamically in AD as CSF Aβ (Roher et al., 2009; Palmqvist et al., 2019). This is highlighted in the AlzBiomarker database, which reveals that in contrast to the decreases in CSF Aβ concentrations, which is expected as this reflects Aβ aggregation into plaques, plasma Aβ1–40 increases by 4%, and plasma Aβ1–42 increases by 5%. The marginal increases in these biomarkers in plasma, as well as the lower overall degree of change observed, is most likely a consequence of the peripheral production of Aβ, which is unaffected by pathology. This highlights the need to develop and measure biomarkers of Aβ pathology which can better distinguish between AD and non-AD individuals, particularly in blood, but which also better reflect the pathology occurring within the brains of AD patients. This is where looking at the markers of tissue reaction to Aβ may prove useful.

Markers of Tissue Reaction to Amyloid-β

The deposition of Aβ within the brain parenchyma results in numerous effects on the surrounding tissue, including glial cell activation and neuroinflammation, tau phosphorylation, and neurodegeneration, as already discussed. Increasing evidence is showing that it may be possible to use markers of these processes as indirect markers of cerebral Aβ pathology.

Glial Fibrillary Acidic Protein

GFAP is a well-known marker of astrocytosis in the CNS. Early in vivo studies observed Aβ-containing astrocytes in the brains of AD patients (Funato et al., 1998; Thal et al., 2000). A subsequent in vitro investigation revealed that astrocytes can phagocytose Aβ (Wyss-Coray et al., 2003), which is the most likely cause of the intracellular Aβ observed in the two aforementioned in vivo studies. Whilst the exact role of astrocytosis in AD remains unclear, it is apparent that reactive astrocytes follow the same spatial distribution as plaques in post-mortem analyses of AD brains (Beach and McGeer, 1988; Perez-Nievas and Serrano-Pozo, 2018). Furthermore, investigations have revealed that reactive astrocytes are involved in Aβ production and toxicity (Garwood et al., 2011; Söllvander et al., 2016). It was previously thought that the number of astrocytes surrounding plaques increases as the disease progresses (Pike et al., 1995; Vehmas et al., 2003). However, more recent studies using a combination of PET tracers have revealed that astrocytosis (depicted by the 11C-deuterenium-L-deprenyl tracer), is an early phenomenon in AD (depicted by the 11C-Pittsburgh compound-B tracer for Aβ plaques), and this astrocytosis decreases as plaque load increases (Carter et al., 2012; Scholl et al., 2015; Rodriguez-Vieitez et al., 2016).

Studies have shown that CSF GFAP concentrations in AD are significantly increased compared to healthy controls (Colangelo et al., 2014; Ishiki et al., 2016), and are significantly increased in the cognitively unimpaired Aβ-positive, tau-positive preclinical stage of AD (Milà-Alomà et al., 2020). However, cross-disease comparisons between AD, frontotemporal lobe dementia (FTLD) and dementia with Lewy bodies (DLB) reveal that CSF GFAP concentrations are significantly increased in all three diseases compared to controls, with FTLD concentrations being significantly greater compared to AD and DLB (Ishiki et al., 2016). This highlights that elevated CSF GFAP is not specific to AD, and hence has little diagnostic value in distinguishing AD from other neurodegenerative diseases.

Interest in GFAP as a plasma biomarker for AD came about due to the possibility of more sensitive assays making it possible to measure within blood. Similar to CSF GFAP, elevated plasma GFAP concentrations have been observed in a variety of neurodegenerative and non-neurodegenerative neurological conditions, including AD (Mayer et al., 2013; Elahi et al., 2020; Heller et al., 2020; van Ballegoij et al., 2020). However, further investigations have revealed that plasma GFAP concentrations correlate strongly with cerebral Aβ pathology, as measured by PET (Verberk et al., 2020), as well as with decreasing white matter volume and worsening cognitive function (Oeckl et al., 2019; Rajan et al., 2020; Verberk et al., 2020; Asken et al., 2021), and hence it is relatively Aβ-specific. In fact, simultaneous comparisons in two independent cohorts between plasma GFAP and NfL, a sensitive biomarker of neuronal injury independent of Aβ pathology, revealed that plasma GFAP may be more sensitive to cortical and cognitive changes than plasma NfL (Asken et al., 2021). Plasma GFAP is higher in Aβ-positive cognitively unimpaired individuals at risk of developing AD (Chatterjee et al., 2021), and longitudinal investigations have observed that plasma GFAP can predict subsequent conversion of mild cognitive impairment (MCI) patients to AD with an area under the receiver operating characteristic curve of 0.84 (95% CI 0.77–0.91) (Cicognola et al., 2021). Furthermore, individuals with a positive CSF Aβ1–42/1–40, but with Aβ PET levels below the cut off for being deemed Aβ PET-positive (i.e., individuals in the earliest preclinical stage of AD), were observed to have significantly higher plasma GFAP concentrations than Aβ-negative individuals, despite there being no significant difference in CSF GFAP concentrations between the two groups (Benedet et al., 2021). One possible reason for this is that GFAP may be released more directly into the bloodstream by astrocytic end feet, thus making plasma changes in GFAP concentrations more pronounced than changes in CSF GFAP concentrations (Giannoni et al., 2018). This is further supported by a plethora of evidence highlighting that the integrity of the BBB is abnormal in AD, resulting in microvascular leakage of proteins into the blood (Banks, 2012). Another reason for significant increases in plasma GFAP, but not CSF GFAP, in AD patients may be due to GFAP being extremely stable in blood, whereas CSF GFAP is much more sensitive to freeze-thaw cycles overtime (Abdelhak et al., 2019; Ashton et al., 2021). However, further work must be undertaken to better understand the reason for this discrepancy between plasma and CSF GFAP. Nonetheless, together these studies highlight that astrocytosis begins in the prodromal stage of AD, and elevated plasma GFAP is associated with neuronal injury, worsening cognition, and markers of cerebral Aβ pathology. Whilst plasma Aβ1–42/1–40 may also function well as a plasma biomarker of Aβ pathology, plasma GFAP appears to give a much broader picture of the state of the individual, hence it may better function as a surrogate plasma biomarker for Aβ pathology in AD. This is further supported by a 50% increase in CSF GFAP compared to a 93% increase in plasma (see text footnote 1; Accessed 7th December 2021) in AD vs controls, with only the plasma GFAP changes being statistically significant.

Triggering Receptor Expressed on Myeloid Cells 2

As discussed, neuroinflammation is emerging as a central player in AD pathogenesis and neuropathology (Bronzuoli et al., 2016). In response to Aβ, microglia upregulate the expression of TREM2, an innate immune transmembrane receptor expressed by myeloid cells. Within the CNS, TREM2 is unique to microglia, however, peripherally it is also expressed by immature dendritic cells and osteoclasts (Neumann and Takahashi, 2006; Jiang et al., 2014; Ghosh et al., 2021). TREM2 is required to initiate and promote microglial activation and Aβ phagocytosis (Heneka et al., 2015; Wang et al., 2015), and it plays an essential role in maintaining CNS homeostasis by mediating the phagocytic function of microglia, suppressing pro-inflammatory cytokine release and enhancing transcription of anti-inflammatory cytokines (Neumann and Takahashi, 2006; Paradowska-Gorycka and Jurkowska, 2013). This is further highlighted by a homozygous loss of function mutation in the TREM2-encoding gene resulting in a neurological disease characterised by: (1) chronic neurodegeneration, most likely caused by the decreased clearance of debris by microglia, and a resultant increase in proinflammatory cytokines and neuroinflammation; and (2) bone cysts due to abnormal osteoclast maturation and function (Paloneva et al., 2001, 2002; Neumann and Takahashi, 2006). Furthermore, early AD studies have revealed that activated microglia concentrate around Aβ deposits in both AD patients and mouse models of AD, highlighting the role receptors must play in mediating this interaction (Dickson et al., 1993; Paresce et al., 1996; Frautschy et al., 1998). In particular, the discovery that a heterozygous missense mutation in the TREM2-encoding gene is a risk factor for development of AD, to a similar extent as that observed for apolipoprotein ε4, highlights the role TREM2, and hence microglia, play in AD neuropathology (Guerreiro et al., 2013; Jonsson et al., 2013; Slattery et al., 2014; Rosenthal et al., 2015).

When TREM2 is proteolytically cleaved, sTREM2 is produced and can be measured in CSF to assess TREM2 activity, and hence act as a surrogate marker for microglial activation (Ulrich et al., 2017). Attempts to measure CSF sTREM2 concentrations in AD have shown significantly higher concentrations in pre-symptomatic AD, MCI, and early/mild AD (Henjum et al., 2016; Heslegrave et al., 2016; Piccio et al., 2016; Suarez-Calvet et al., 2016). As the disease progresses, CSF sTREM2 concentrations appear to decrease, such that there is either no significant difference, or a significant reduction, in AD compared to controls (Kleinberger et al., 2014; Henjum et al., 2016; Suarez-Calvet et al., 2016). This highlights a longitudinal change in CSF sTREM2, with concentrations peaking in MCI and progressively decreasing as disease severity increases (Suarez-Calvet et al., 2016). Interestingly, CSF sTREM2 concentrations have been found to correlate with CSF total tau (t-tau) and p-tau, but not with CSF Aβ1–42 (Heslegrave et al., 2016; Piccio et al., 2016). This suggests that although TREM2 correlates with neuronal injury and tau pathology markers, it may not reflect Aβ pathology, which is further highlighted by elevated CSF sTREM2 levels in the absence of Aβ pathology (Suarez-Calvet et al., 2016). Evidence has shown that Aβ deposition and neuronal injury precede elevated CSF sTREM2 concentrations in pre-symptomatic patients (Suárez-Calvet et al., 2016). Therefore, it is possible that as the disease progresses, there is a failure of microglial function, particularly in phagocytosing Aβ plaques, and hence reduced TREM2 activity. Alternatively, TREM2 may be a marker of Aβ-induced tau pathology and neurodegeneration in AD (Park et al., 2021), supporting the notion that microglial activation may drive tau pathology (Pascoal et al., 2021).

Unlike CSF sTREM2, where there is a 31% increase in AD compared to controls (see text footnote 1; Accessed 7th December 2021), no significant difference has been observed in plasma sTREM2 concentrations (Kleinberger et al., 2014; Piccio et al., 2016; Bekris et al., 2018). Furthermore, plasma sTREM2 concentrations do not correlate with CSF t-tau, p-tau, or Aβ1–42 (Piccio et al., 2016; Bekris et al., 2018). Interestingly, Bekris et al. (2018) observed a significant positive correlation between plasma and CSF sTREM2, whilst Piccio et al. (2016) observed a non-significant positive correlation between plasma and CSF sTREM2 concentrations, and Park et al. (2021) observed a negative correlation between plasma and CSF sTREM2. The cause of the differing results between these studies is not clear but may be due to differences in cohort sizes or methodological factors. A longitudinal study found elevated serum sTREM2 constituted an increased risk of dementia (Ohara et al., 2019). However, the criteria used by Ohara et al. (2019) to diagnose AD within their patient cohort were purely clinical, with no biomarker evidence of AD assessed, therefore it is possible that some patients were misdiagnosed with AD. Whilst sTREM2 is measurable in both plasma and serum, direct comparisons of absolute levels in these matrices, to our knowledge, have not been performed. Overall, the general consensus appears to be that plasma sTREM2 may not be a useful biomarker for use in AD, particularly with regards to tissue reactions to Aβ. This is because plasma sTREM2 can reflect peripheral inflammation produced by other cells of myeloid origin, in contrast to CSF sTREM2, which is microglia-specific (Rodriguez-Vieitez and Ashton, 2021).

Phosphorylated Tau

Similar to Aβ, tau is a natural component of mature neurones, with some healthy individuals also having a small percentage of p-tau, as phosphorylation appears to be important in enabling the normal function of tau within neurones (Barthélemy et al., 2020). However, in AD, tau is 3–4 fold more phosphorylated, and it is this hyperphosphorylation that promotes the intraneuronal aggregation of tau into NFTs (Vanmechelen et al., 2000; Parnetti et al., 2001; Buerger et al., 2002). However, as well as aggregating intraneuronally, p-tau is secreted from neurones, and can subsequently be measured in CSF and blood. In fact, it is possible that CSF changes in p-tau occur prior to NFT formation (Barthélemy et al., 2020). There are up to 85 sites at which tau can be phosphorylated (Martin et al., 2011), with the three most widely investigated sites in relation to AD being tau phosphorylated at threonine 181 (p-tau181), threonine 217 (p-tau217), and threonine 231 (p-tau231). In contrast to t-tau, which is included in the AT(N) criteria for AD diagnosis as a general marker of neurodegeneration and neuronal injury, there is no change in CSF p-tau concentrations in other tauopathies and neurological conditions (Hesse et al., 2001; Parnetti et al., 2001; Riemenschneider et al., 2003). Rather, CSF p-tau is significantly increased in AD compared to controls and other neurodegenerative diseases, regardless of which epitope is measured (Kohnken et al., 2000; Vanmechelen et al., 2000; Buerger et al., 2002; Verbeek et al., 2004; Welge et al., 2009). However, when combined with increased CSF p-tau, increased CSF t-tau does indeed reflect AD pathology, as opposed to being a general indication of neuronal injury (Alawode et al., 2021). Comparing and contrasting p-tau181, p-tau217, and p-tau231 is beyond the scope of this review, so we will look at p-tau in general. However, the analytical and clinical performance of assays detecting these three tau epitopes was recently assessed by Bayoumy et al. (2021).

In light of CSF p-tau being significantly increased in AD compared to controls, several studies have shown a clear correlation between CSF p-tau and Aβ pathology measures (Bateman et al., 2012; Barthélemy et al., 2020; Suárez-Calvet et al., 2020), with changes in CSF p-tau also being observed several years prior to symptom onset, and when only subtle changes in Aβ pathology measures are detected. Furthermore, CSF p-tau has been shown to correlate more strongly with cognitive impairment than Aβ biomarkers (Gómez-Isla et al., 1997; Nelson et al., 2012; Jack et al., 2018). Given that increases in CSF p-tau are unique to AD, and are not observed in other tauopathies, it is hypothesised that p-tau may be a measure of Aβ-induced tau phosphorylation (Maia et al., 2013; Kanmert et al., 2015; Sato et al., 2018). Furthermore, CSF p-tau181, p-tau217, and p-tau231 exhibit remarkably high increases of 87, 999, and 489%, respectively, in AD compared to controls (see text footnote 1; Accessed 7th December 2021). Given the overwhelming evidence showing CSF p-tau to be a robust biomarker for AD, the question lies with whether plasma p-tau correlates as strongly with Aβ pathology, and whether there is scope for plasma p-tau to function as a biomarker of Aβ pathology better than Aβ biomarkers.

Attempts to quantify plasma p-tau began in 2016 and have proven largely successful. Similar to CSF p-tau, plasma p-tau has been found to increase in AD compared to MCI, non-AD dementias and cognitively unimpaired controls (Shekhar et al., 2016; Tatebe et al., 2017; Janelidze et al., 2020; Karikari et al., 2020b). In particular, Mielke et al. (2018) showed that plasma p-tau is strongly associated with Aβ PET imaging, and is highly sensitive and specific to increased cerebral Aβ burden, whilst Karikari et al. (2020a) showed that plasma p-tau increases markedly in Aβ PET negative individuals who also have decreased CSF Aβ concentrations. Furthermore, in their cross-sectional study looking at biomarker trajectories with increasing Aβ burden, Palmqvist et al. (2019) showed that in AD plasma p-tau changes significantly before CSF and plasma Aβ1–42/1–40, and CSF p-tau, all of which exhibit changes before Aβ PET positivity is detected. In addition, they showed that plasma p-tau continues to increase as Aβ burden increases (Palmqvist et al., 2019). This highlights that plasma p-tau may be one of the earliest biomarkers to change in AD, and it continues to reflect Aβ pathology whilst also giving additional information on the progression of tau pathology up to 10 years before tau PET positivity is detected (Bateman et al., 2012). This is further highlighted by increases in plasma p-tau181 and p-tau217 of 80 and 288%, respectively, in AD compared to controls (see text footnote 1; Accessed 7th December 2021).

Neurofilament Light Chain

Along with microtubules and microfilaments, neurofilaments form the neuronal cytoskeleton (Yuan and Nixon, 2021). However, one particular subunit, NfL, is expressed predominantly in large-calibre myelinated axons (Schlaepfer and Lynch, 1977). Following neuronal damage and degeneration, NfL is released into the extracellular space, and is detectable both in CSF and in blood (Lista et al., 2017). Therefore, by proxy, biofluid changes in NfL are not specific to AD, but reflect general neuronal death and axonal loss. Nonetheless, CSF NfL is significantly increased in AD compared to controls, and predicts progression from MCI to AD (Sjögren et al., 2001; Petzold et al., 2007; Mattsson et al., 2016; Olsson et al., 2016; Zetterberg et al., 2016; Lista et al., 2017). Of particular interest for this review is whether CSF NfL correlates with Aβ pathology. A study by Zetterberg et al. (2016) observed that whilst there were correlations between increased CSF NfL and decreased CSF Aβ1–42, there was no significant difference in CSF NfL concentrations between the Aβ-positive and Aβ-negative groups. This has been further corroborated by several studies (Jin et al., 2019; Aschenbrenner et al., 2020; Dhiman et al., 2020). Interestingly, Dhiman et al. (2020) observed that a combination of CSF NfL and a ratio between NfL and Aβ1–42 (NfL/Aβ1–42) predicted Aβ burden, brain atrophy and altered cognition. Nonetheless, these studies highlight that changes in CSF NfL occur independently of Aβ pathology, therefore CSF NfL is not suitable as a surrogate biomarker for Aβ pathology in AD. Rather, evidence suggests that CSF NfL correlates better with tau biomarkers (Olsson et al., 2016; Zetterberg et al., 2016; Dhiman et al., 2020).

Overwhelming evidence has shown that not only does plasma NfL significantly increase in AD compared to controls, but it also correlates with CSF NfL and tau biomarkers (Mattsson et al., 2017; Lewczuk et al., 2018; Lin et al., 2018). Mattsson et al. (2017) and Lewczuk et al. (2018) both observed a correlation between increased plasma NfL and decreased CSF Aβ1–42. However, when this correlation was investigated further, Lewczuk et al. (2018) found it to no longer be significant when the diagnosis of each individual was taken into consideration – a finding supported by Sanchez-Valle et al. (2018). Although increases in plasma NfL are not unique to AD, and can be seen in several other neurodegenerative and non-neurodegenerative conditions (Gisslén et al., 2016; Rohrer et al., 2016; Rojas et al., 2016; Weydt et al., 2016), a recent longitudinal study revealed that plasma NfL is increased up to 22 years prior to expected AD symptom onset (Quiroz et al., 2020), which is consistent with earlier studies of serum NfL in FAD (Preische et al., 2019; Weston et al., 2019). With very little difference between plasma and CSF NfL increases in AD compared to controls (98% in CSF vs 85% in plasma; see text footnote 1; Accessed 7th December 2021) it is clear that plasma NfL may play a more advantageous role in AD than CSF NfL. However, neither plasma nor CSF NfL measures correlate with Aβ neuropathology, hence are not suitable for use as surrogate biomarkers for Aβ.

Conclusion

In this manuscript, we have considered Aβ as one of the causes of AD, as described in the amyloid cascade hypothesis, and how this can be detected and monitored using biofluid-based biomarkers. Furthermore, we have investigated the evidence supporting and opposing four key biomarkers of tissue reactions to Aβ being used as indirect biomarkers of Aβ pathology in AD diagnosis and clinical trials – GFAP, TREM2, p-tau, and NfL. Up until now, Aβ PET was the gold standard for confirming the presence of Aβ pathology in clinical trials. CSF Aβ measurements have recently been used in conjunction with Aβ PET and have proven to detect pathology at an earlier stage. However, we now have access to sensitive tests for plasma Aβ, which have revolutionised the field. There is now a move towards blood-based biomarker measurements in AD due to blood being cheaper and less invasive to obtain. However, the peripheral production of Aβ makes measuring this protein in blood, and distinguishing between AD, non-AD and controls, particularly challenging, as there is significant overlap in plasma Aβ concentrations between these groups. Therefore, finding an alternative biomarker to measure Aβ pathology indirectly is extremely advantageous for improving diagnostic accuracy, monitoring disease progression, and assessing response to treatment. With regards to sensitivity and specificity, for some biomarkers, including Aβ1–42/1–40, diagnostic accuracy for CSF measurement is much greater than that of blood measurements (Palmqvist et al., 2020). Whereas, some biomarkers perform as well in blood as in CSF, such as p-tau217 (Palmqvist et al., 2020).

Of the four tissue response biomarkers discussed, plasma GFAP appears to be the most promising indirect biomarker of Aβ pathology. In particular, it gives a broader picture of the pathological state of the individual, also providing additional information on neuronal injury and cognitive decline, as well as highlighting astrocyte response to AD pathology. As such, it would be a useful biomarker for preliminary AD diagnosis, and in clinical trials for pre-screening purposes. Furthermore, in relation to drugs targeting Aβ, plasma GFAP could provide useful information on drug efficacy and therapy response, as if Aβ is decreasing successfully with the use of the drug, the tissue reaction to Aβ should also decrease. Whilst plasma p-tau also reflects Aβ pathology, and it does so earlier than plasma and CSF Aβ measures, plasma p-tau would serve better as an early marker of tau pathology, in particular to confirm the presence of Aβ-induced tau phosphorylation. Additionally, it could function well as a therapy response marker. In contrast, plasma sTREM2 shows no significant difference in AD compared to controls, so has no evident role in AD diagnosis and clinical trials at this stage. Finally, whilst plasma NfL does correlate with markers of tau pathology and neurodegeneration, it shows no correlation with markers of Aβ pathology. Nonetheless, it is an excellent biomarker of neuronal injury, so could function in combination with other AD pathology markers to confirm the presence of neurodegeneration, and as a dynamic biomarker, it can also be used to monitor treatments.

One thing that has not been discussed is the role these biomarkers may play in personalised medicine approaches to AD treatment. Whilst the breadth of licensed drugs for AD treatment is currently relatively small, the biomarkers of tissue reactions to Aβ could give further insight into which disease pathways appear to be most active in individual patients, which in itself will provide valuable information on which disease pathways should be specifically targeted within that patient. This may pave the way for a repurposing of drugs currently used to treat other non-AD pathologies. It is clear that biomarkers of tissue reactions to Aβ have a role to play in AD research and clinical practice, whether that be in diagnosis, clinical trials and/or treatment. The recent advances in measurements of blood biomarkers that reflect brain pathology, including Aβ-specific reactions, will enable large scale screening for trial recruitment, meaning that a move towards blood-based biomarkers for AD will be key for this rapidly changing field.

Author Contributions

DA wrote the manuscript. All authors contributed to the concept and content of review, manuscript revision, read, and approved the final version.

Funding

DA was supported by the International Journal of Experimental Pathology and the UK Dementia Research Institute at UCL. NF acknowledges support from the National Institute for Health Research University College London Hospitals Biomedical Research Centre, Rosetrees Trust, Alzheimer’s Research UK, and the UK Dementia Research Institute at UCL. HZ was a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712), Swedish State Support for Clinical Research (#ALFGBG-720931), the Alzheimer Drug Discovery Foundation (ADDF), United States (#201809-2016862), the AD Strategic Fund and the Alzheimer’s Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2019-0228), the European Union’s Horizon 2020 Research and Innovation Programme under the Marie Skłodowska-Curie grant agreement no. 860197 (MIRIADE), European Union Joint Program for Neurodegenerative Disorders (JPND2021-00694), and the UK Dementia Research Institute at UCL.

Conflict of Interest

NF served as a consultant, at advisory boards, or on a data monitoring committee for Roche, Biogen, and Ionis. HZ served at scientific advisory boards and/or as a consultant for Abbvie, Alector, Annexon, Artery Therapeutics, AZTherapies, CogRx, Denali, Eisai, Nervgen, Pinteon Therapeutics, Red Abbey Labs, Passage Bio, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave, has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure, Biogen, and Roche, and was a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

Aβ, amyloid beta; AD, Alzheimer’s disease; APP, amyloid beta precursor protein; AT(N), amyloid, tau, (neurodegeneration); CNS, central nervous system; CSF, cerebrospinal fluid; DLB, dementia with Lewy bodies; DS, down syndrome; ELISA, enzyme-linked immunosorbent assay; FAD, familial Alzheimer’s disease; FTLD, frontotemporal lobe dementia; GFAP, glial fibrillary acidic protein; MCI, mild cognitive impairment; MS, mass spectrometry; NfL, neurofilament light chain; NFT, neurofibrillary tangles; PET, positron emission tomography; p-tau, phosphorylated tau; Simoa, Single molecule array; sTREM2, soluble triggering receptor expressed on myeloid cells 2; TREM2, triggering receptor expressed on myeloid cells 2.

Footnotes

  1. ^ https://www.alzforum.org/alzbiomarker/ad-vs-ctrl

References

Abdelhak, A., Hottenrott, T., Morenas-Rodríguez, E., Suárez-Calvet, M., Zettl, U. K., Haass, C., et al. (2019). Glial activation markers in csf and serum from patients with primary progressive multiple sclerosis: potential of serum GFAP as Disease Severity Marker? Front. Neurol. 10:280. doi: 10.3389/fneur.2019.00280

PubMed Abstract | CrossRef Full Text | Google Scholar

Alawode, D. O. T., Heslegrave, A. J., Ashton, N. J., Karikari, T. K., Simrén, J., Montoliu-Gaya, L., et al. (2021). Transitioning from cerebrospinal fluid to blood tests to facilitate diagnosis and disease monitoring in Alzheimer’s disease. J. Intern. Med. 290, 583–601. doi: 10.1111/joim.13332

PubMed Abstract | CrossRef Full Text | Google Scholar

Albani, D., Marizzoni, M., Ferrari, C., Fusco, F., Boeri, L., Raimondi, I., et al. (2019). Plasma A beta(42) as a biomarker of prodromal Alzheimer’s disease progression in patients with amnestic mild cognitive impairment: evidence from the PharmaCog/E-ADNI Study. J. Alzheimers Dis. 69, 37–48. doi: 10.3233/JAD-180321

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez, A., Toro, R., Cáceres, A., and Maccioni, R. B. (1999). Inhibition of tau phosphorylating protein kinase cdk5 prevents β-amyloid-induced neuronal death. FEBS Lett. 459, 421–426. doi: 10.1016/s0014-5793(99)01279-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez, R., Alvarez, V., Lahoz, C. H., Martínez, C., Peña, J., Sánchez, J. M., et al. (1999). Angiotensin converting enzyme and endothelial nitric oxide synthase DNA polymorphisms and late onset Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry 67, 733–736. doi: 10.1136/jnnp.67.6.733

PubMed Abstract | CrossRef Full Text | Google Scholar

Aschenbrenner, A. J., Gordon, B. A., Fagan, A. M., Schindler, S. E., Balota, D. A., Morris, J. C., et al. (2020). Neurofilament light predicts decline in attention but not episodic memory in preclinical Alzheimer disease. J. Alzheimers Dis. 74, 1119–1129. doi: 10.3233/jad-200018

PubMed Abstract | CrossRef Full Text | Google Scholar

Ashton, N. J., Suárez-Calvet, M., Karikari, T. K., Lantero-Rodriguez, J., Snellman, A., Sauer, M., et al. (2021). Effects of pre-analytical procedures on blood biomarkers for Alzheimer’s pathophysiology, glial activation, and neurodegeneration. Alzheimers Dement. 13:e12168. doi: 10.1002/dad2.12168

PubMed Abstract | CrossRef Full Text | Google Scholar

Asken, B. M., Vandevrede, L., Rojas, J. C., Fonseca, C., Staffaroni, A. M., Elahi, F. M., et al. (2021). Lower white matter volume and worse executive functioning reflected in higher levels of plasma GFAP among Older Adults with and Without Cognitive Impairment. J. Int. Neuropsychol. Soc. doi: 10.1017/S1355617721000813 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Banks, W. A. (2012). Drug delivery to the brain in Alzheimer’s disease: consideration of the blood–brain barrier. Adv. Drug Deliv. Rev. 64, 629–639. doi: 10.1016/j.addr.2011.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Barthélemy, N. R., Li, Y., Joseph-Mathurin, N., Gordon, B. A., Hassenstab, J., Benzinger, T. L. S., et al. (2020). A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nat. Med. 26, 398–407. doi: 10.1038/s41591-020-0781-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Bateman, R. J., Xiong, C., Benzinger, T. L. S., Fagan, A. M., Goate, A., Fox, N. C., et al. (2012). Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N. Engl. J. Med. 367, 795–804. doi: 10.1056/NEJMoa1202753

PubMed Abstract | CrossRef Full Text | Google Scholar

Bayoumy, S., Verberk, I. M. W., Den Dulk, B., Hussainali, Z., Zwan, M., Van Der Flier, W. M., et al. (2021). Clinical and analytical comparison of six Simoa assays for plasma P-tau isoforms P-tau181, P-tau217, and P-tau231. Alzheimers Res. Ther. 13:198.

Google Scholar

Beach, T. G., and McGeer, E. G. (1988). Lamina-specific arrangement of astrocytic gliosis and senile plaques in Alzheimer’s disease visual cortex. Brain Res. 463, 357–361. doi: 10.1016/0006-8993(88)90410-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Behl, C., Davis, J. B., Klier, F. G., and Schubert, D. (1994). Amyloid β peptide induces necrosis rather than apoptosis. Brain Res. 645, 253–264. doi: 10.1016/0006-8993(94)91659-4

CrossRef Full Text | Google Scholar

Bekris, L. M., Khrestian, M., Dyne, E., Shao, Y., Pillai, J. A., Rao, S. M., et al. (2018). Soluble TREM2 and biomarkers of central and peripheral inflammation in neurodegenerative disease. J. Neuroimmunol. 319, 19–27. doi: 10.1016/j.jneuroim.2018.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Benedet, A. L., Milà-Alomà, M., Vrillon, A., Ashton, N. J., Pascoal, T. A., Lussier, F., et al. (2021). Differences between plasma and cerebrospinal fluid glial Fibrillary acidic protein levels across the alzheimer disease continuum. JAMA Neurol. 78, 1471–1483. doi: 10.1001/jamaneurol.2021.3671

PubMed Abstract | CrossRef Full Text | Google Scholar

Blennow, K. (2021). Plasma pTau isoform assessment across disease stages. Alzheimers Dement. 17:e050710.

Google Scholar

Blennow, K., Mattsson, N., Schöll, M., Hansson, O., and Zetterberg, H. (2015). Amyloid biomarkers in Alzheimer’s disease. Trends Pharmacol. Sci. 36, 297–309.

Google Scholar

Blennow, K., and Zetterberg, H. (2015). Understanding biomarkers of neurodegeneration: ultrasensitive detection techniques pave the way for mechanistic understanding. Nat. Med. 21, 217–219. doi: 10.1038/nm.3810

PubMed Abstract | CrossRef Full Text | Google Scholar

Bronzuoli, M. R., Iacomino, A., Steardo, L., and Scuderi, C. (2016). Targeting neuroinflammation in Alzheimer’s disease. J. Inflamm. Res. 9, 199–208. doi: 10.2147/jir.s86958

PubMed Abstract | CrossRef Full Text | Google Scholar

Bruggink, K. A., Bea Kuiperij, H., Ahr Claassen, J., and Verbeek, M. (2013). The diagnostic value of CSF amyloid-β43 in differentiation of dementia syndromes. Curr. Alzheimer Res. 10, 1034–1040. doi: 10.2174/15672050113106660168

PubMed Abstract | CrossRef Full Text | Google Scholar

Buerger, K., Zinkowski, R., Teipel, S. J., Tapiola, T., Arai, H., Blennow, K., et al. (2002). Differential diagnosis of Alzheimer disease with cerebrospinal fluid levels of Tau protein phosphorylated at threonine 231. Arch. Neurol. 59, 1267–1272. doi: 10.1001/archneur.59.8.1267

PubMed Abstract | CrossRef Full Text | Google Scholar

Busciglio, J., Lorenzo, A., Yeh, J., and Yankner, B. A. (1995). β-Amyloid fibrils induce tau phosphorylation and loss of microtubule binding. Neuron 14, 879–888. doi: 10.1016/0896-6273(95)90232-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Cacace, R., Sleegers, K., and Van Broeckhoven, C. (2016). Molecular genetics of early-onset Alzheimer’s disease revisited. Alzheimers Dement. 12, 733–748. doi: 10.1016/j.jalz.2016.01.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, Z., Hussain, M. D., and Yan, L.-J. (2014). Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int. J. Neurosci. 124, 307–321. doi: 10.3109/00207454.2013.833510

PubMed Abstract | CrossRef Full Text | Google Scholar

Cameron, B., and Landreth, G. E. (2009). Inflammation, microglia, and Alzheimer’s disease. Neurobiol. Dis. 37, 503–509. doi: 10.1016/j.nbd.2009.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Canevari, L., Abramov, A. Y., and Duchen, M. R. (2004). Toxicity of Amyloid β peptide: tales of calcium, mitochondria, and oxidative stress. Neurochem. Res. 29, 637–650. doi: 10.1023/b:nere.0000014834.06405.af

PubMed Abstract | CrossRef Full Text | Google Scholar

Carter, S. F., Scholl, M., Almkvist, O., Wall, A., Engler, H., Langstrom, B., et al. (2012). Evidence for astrocytosis in prodromal alzheimer disease provided by C-11-Deuterium-L-Deprenyl: a multitracer PET paradigm combining C-11-pittsburgh compound B and F-18-FDG. J. Nucl. Med. 53, 37–46. doi: 10.2967/jnumed.110.087031

PubMed Abstract | CrossRef Full Text | Google Scholar

Chatani, E., and Yamamoto, N. (2018). Recent progress on understanding the mechanisms of amyloid nucleation. Biophys. Rev. 10, 527–534. doi: 10.1007/s12551-017-0353-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Chatterjee, P., Peini, S., Stoops, E., Goozee, K., Villemagne, V. L., Asih, P. R., et al. (2021). Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer’s disease. Transl. Psychiatry 11:68. doi: 10.1038/s41398-020-01137-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, G.-F., Xu, T.-H., Yan, Y., Zhou, Y.-R., Jiang, Y., Melcher, K., et al. (2017). Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol. Sin. 38, 1205–1235. doi: 10.1038/aps.2017.28

PubMed Abstract | CrossRef Full Text | Google Scholar

Cicognola, C., Janelidze, S., Hertze, J., Zetterberg, H., Blennow, K., Mattsson-Carlgren, N., et al. (2021). Plasma glial fibrillary acidic protein detects Alzheimer pathology and predicts future conversion to Alzheimer dementia in patients with mild cognitive impairment. Alzheimers Res. Ther. 13:68. doi: 10.1186/s13195-021-00804-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Cohen, L., and Walt, D. R. (2019). Highly sensitive and multiplexed protein measurements. Chem. Rev. 119, 293–321. doi: 10.1021/acs.chemrev.8b00257

PubMed Abstract | CrossRef Full Text | Google Scholar

Colangelo, A. M., Alberghina, L., and Papa, M. (2014). Astrogliosis as a therapeutic target for neurodegenerative diseases. Neurosci. Lett. 565, 59–64. doi: 10.1016/j.neulet.2014.01.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Colin, L. M., Gail, S., Nicola, A. W., Gerd, M., Brian, L. M., and Konrad, B. (1985). Amyloid plaque core protein in Alzheimer Disease and down syndrome. Proc. Natl. Acad. Sci. U.S.A. 82, 4245–4249. doi: 10.1073/pnas.82.12.4245

PubMed Abstract | CrossRef Full Text | Google Scholar

Crutchfield, C. A., Thomas, S. N., Sokoll, L. J., and Chan, D. W. (2016). Advances in mass spectrometry-based clinical biomarker discovery. Clin. Proteomics 13:1. doi: 10.1186/s12014-015-9102-9

PubMed Abstract | CrossRef Full Text | Google Scholar

DeKosky, S. T. (2001). Epidemiology and pathophysiology of Alzheimer’s disease. Clin. Cornerstone 3, 15–26.

Google Scholar

DeMattos, R. B., Lu, J., Tang, Y., et al. (2012). A plaque-specific antibody clears existing β-amyloid Plaques in Alzheimer’s Disease mice. Neuron 76, 908–920. doi: 10.1016/j.neuron.2012.10.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Deture, M. A., and Dickson, D. W. (2019). The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 14:32.

Google Scholar

Dhiman, K., Gupta, V. B., Villemagne, V. L., Eratne, D., Graham, P. L., Fowler, C., et al. (2020). Cerebrospinal fluid neurofilament light concentration predicts brain atrophy and cognition in Alzheimer’s disease. Alzheimer’s Dement. 12:e12005. doi: 10.1002/dad2.12005

PubMed Abstract | CrossRef Full Text | Google Scholar

Dickson, D. W., Lee, S. C., Mattiace, L. A., Yen, S.-H. C., and Brosnan, C. (1993). Microglia and cytokines in neurological disease, with special reference to AIDS and Alzheimer’s disease. Glia 7, 75–83. doi: 10.1002/glia.440070113

PubMed Abstract | CrossRef Full Text | Google Scholar

Elahi, F. M., Casaletto, K. B., La Joie, R., Walters, S. M., Harvey, D., Wolf, A., et al. (2020). Plasma biomarkers of astrocytic and neuronal dysfunction in early- and late-onset Alzheimer’s disease. Alzheimer’s Dement. 16, 681–695. doi: 10.1016/j.jalz.2019.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Finder, V. H., and Glockshuber, R. (2007). Amyloid-β aggregation. Neurodegener. Dis. 4, 13–27.

Google Scholar

Frautschy, S. A., Yang, F., Irrizarry, M., Hyman, B., Saido, T. C., Hsiao, K., et al. (1998). Microglial response to amyloid plaques in APPsw transgenic mice. Am. J. Pathol. 152, 307–317.

PubMed Abstract | Google Scholar

Funato, H., Yoshimura, M., Yamazaki, T., Saido, T. C., Ito, Y., Yokofujita, J., et al. (1998). Astrocytes containing amyloid beta-protein (A beta)-positive granules are associated with AP40-positive diffuse plaques in the aged human brain. Am. J. Pathol. 152, 983–992.

Google Scholar

Galasko, D. (2005). Biomarkers for Alzheimer’s disease - Clinical needs and application. J. Alzheimers Dis. 8, 339–346. doi: 10.3233/jad-2005-8403

PubMed Abstract | CrossRef Full Text | Google Scholar

Garwood, C. J., Pooler, A. M., Atherton, J., Hanger, D. P., and Noble, W. (2011). Astrocytes are important mediators of Aβ-induced neurotoxicity and tau phosphorylation in primary culture. Cell Death Dis. 2:e167. doi: 10.1038/cddis.2011.50

PubMed Abstract | CrossRef Full Text | Google Scholar

Ghosh, P., Singh, R., Ganeshpurkar, A., Pokle, A. V., Singh, R. B., Singh, S. K., et al. (2021). Cellular and molecular influencers of neuroinflammation in Alzheimer’s disease: recent concepts & roles. Neurochem. Int. 151:105212. doi: 10.1016/j.neuint.2021.105212

PubMed Abstract | CrossRef Full Text | Google Scholar

Giannoni, P., Badaut, J., Dargazanli, C., De Maudave, A. F. H., Klement, W., Costalat, V., et al. (2018). The pericyte-glia interface at the blood-brain barrier. Clin. Sci. 132, 361–374. doi: 10.1042/CS20171634

PubMed Abstract | CrossRef Full Text | Google Scholar

Gisslén, M., Price, R. W., Andreasson, U., Norgren, N., Nilsson, S., Hagberg, L., et al. (2016). Plasma concentration of the neurofilament light protein (NFL) is a biomarker of CNS injury in HIV infection: a cross-sectional Study. Ebiomedicine 3, 135–140. doi: 10.1016/j.ebiom.2015.11.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Gómez-Isla, T., Hollister, R., West, H., Mui, S., Growdon, J. H., Petersen, R. C., et al. (1997). Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer’s disease. Ann. Neurol. 41, 17–24. doi: 10.1002/ana.410410106

PubMed Abstract | CrossRef Full Text | Google Scholar

Guerreiro, R., Wojtas, A., Bras, J., Carrasquillo, M., Rogaeva, E., Majounie, E., et al. (2013). TREM2 Variants in Alzheimer’s Disease. New Engl. J. Med. 368, 117–127.

Google Scholar

Guzman-Martinez, L., Maccioni, R. B., Andrade, V., Navarrete, L. P., Pastor, M. G., and Ramos-Escobar, N. (2019). Neuroinflammation as a common feature of neurodegenerative disorders. Front. Pharmacol. 10:1008. doi: 10.3389/fphar.2019.01008

PubMed Abstract | CrossRef Full Text | Google Scholar

Hampel, H., Hardy, J., Blennow, K., Chen, C., Perry, G., Kim, S. H., et al. (2021). The amyloid-β pathway in Alzheimer’s disease. Mol. Psychiatry 26, 5481–5503.

Google Scholar

Hardy, J., and Higgins, G. (1992). Alzheimer’s disease: the amyloid cascade hypothesis. Science 256, 184–185.

Google Scholar

Hardy, J., and Selkoe, D. J. (2002). The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356. doi: 10.1126/science.1072994

PubMed Abstract | CrossRef Full Text | Google Scholar

Harigaya, Y., Saido, T. C., Eckman, C. B., Prada, C.-M., Shoji, M., and Younkin, S. G. (2000). Amyloid β protein starting pyroglutamate at position 3 is a major component of the amyloid deposits in the Alzheimer’s disease brain. Biochem. Biophys. Res. Commun. 276, 422–427. doi: 10.1006/bbrc.2000.3490

PubMed Abstract | CrossRef Full Text | Google Scholar

Hart, N. J., Koronyo, Y., Black, K. L., and Koronyo-Hamaoui, M. (2016). Ocular indicators of Alzheimer’s: exploring disease in the retina. Acta Neuropathol. 132, 767–787. doi: 10.1007/s00401-016-1613-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Heller, C., Foiani, M. S., Moore, K., Convery, R., Bocchetta, M., Neason, M., et al. (2020). Plasma glial fibrillary acidic protein is raised in progranulin-associated frontotemporal dementia. J. Neurol. Neurosurg. Psychiatry 91, 263–270. doi: 10.1136/jnnp-2019-321954

PubMed Abstract | CrossRef Full Text | Google Scholar

Hendricks, R., Baker, D., Brumm, J., Davancaze, T., Harp, C., Herman, A., et al. (2019). Establishment of neurofilament light chain Simoa assay in cerebrospinal fluid and blood. Bioanalysis 11, 1405–1418. doi: 10.4155/bio-2019-0163

PubMed Abstract | CrossRef Full Text | Google Scholar

Heneka, M. T. P., Carson, M. J. P., Khoury, J. E. P., Landreth, G. E. P., Brosseron, F. P., Feinstein, D. L. P., et al. (2015). Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 14, 388–405.

Google Scholar

Henjum, K., Almdahl, I. S., Årskog, V., Minthon, L., Hansson, O., Fladby, T., et al. (2016). Cerebrospinal fluid soluble TREM2 in aging and Alzheimer’s disease. Alzheimers Res. Ther. 8:17. doi: 10.1186/s13195-016-0182-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Heslegrave, A., Heywood, W., Paterson, R., Magdalinou, N., Svensson, J., Johansson, P., et al. (2016). Increased cerebrospinal fluid soluble TREM2 concentration in Alzheimer’s disease. Mol. Neurodegener. 11:3. doi: 10.1186/s13024-016-0071-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Hesse, C., Rosengren, L., Andreasen, N., Davidsson, P., Vanderstichele, H., Vanmechelen, E., et al. (2001). Transient increase in total tau but not phospho-tau in human cerebrospinal fluid after acute stroke. Neurosci. Lett. 297, 187–190. doi: 10.1016/s0304-3940(00)01697-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirayama, A., Horikoshi, Y., Maeda, M., Ito, M., and Takashima, S. (2003). Characteristic developmental expression of amyloid β40, 42 and 43 in patients with Down syndrome. Brain Dev. 25, 180–185. doi: 10.1016/s0387-7604(02)00209-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishiki, A., Kamada, M., Kawamura, Y., Terao, C., Shimoda, F., Tomita, N., et al. (2016). Glial fibrillar acidic protein in the cerebrospinal fluid of Alzheimer’s disease, dementia with Lewy bodies, and frontotemporal lobar degeneration. J. Neurochem. 136, 258–261. doi: 10.1111/jnc.13399

PubMed Abstract | CrossRef Full Text | Google Scholar

Ittner, L. M., and Götz, J. (2011). Amyloid-β and tau — a toxic pas de deux in Alzheimer’s disease. Nat. Rev. Neurosci. 12, 65–72.

Google Scholar

Jack, C. R., Bennett, D. A., Blennow, K., Carrillo, M. C., Dunn, B., Haeberlein, S. B., et al. (2018). NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 14, 535–562. doi: 10.1016/j.jalz.2018.02.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Jack, C. R., Bennett, D. A., Blennow, K., Carrillo, M. C., Feldman, H. H., Frisoni, G. B., et al. (2016). A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 87, 539–547. doi: 10.1212/WNL.0000000000002923

PubMed Abstract | CrossRef Full Text | Google Scholar

Jäkel, L., Boche, D., Nicoll, J. A. R., and Verbeek, M. M. (2019). Aβ43 in human Alzheimer’s disease: effects of active Aβ42 immunization. Acta Neuropathol. Commun. 7:141.

Google Scholar

Janelidze, S., Mattsson, N., Palmqvist, S., Smith, R., Beach, T. G., Serrano, G. E., et al. (2020). Plasma P-tau181 in Alzheimer’s disease: relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s dementia. Nat. Med. 26:379. doi: 10.1038/s41591-020-0755-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Janelidze, S., Stomrud, E., Palmqvist, S., Zetterberg, H., Van Westen, D., Jeromin, A., et al. (2016). Plasma beta-amyloid in Alzheimer’s disease and vascular disease. Sci. Rep. 6:26801. doi: 10.1038/srep26801

PubMed Abstract | CrossRef Full Text | Google Scholar

Janelidze, S., Teunissen, C. E., Zetterberg, H., Allué, J. A., Sarasa, L., Eichenlaub, U., et al. (2021). Head-to-head comparison of 8 plasma amyloid-β 42/40 assays in Alzheimer disease. JAMA Neurol. 78, 1375-1382. doi: 10.1001/jamaneurol.2021.3180

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, T., Tan, L., Zhu, X.-C., Zhang, Q.-Q., Cao, L., Tan, M.-S., et al. (2014). Upregulation of TREM2 ameliorates neuropathology and rescues spatial cognitive impairment in a transgenic mouse model of Alzheimer’s disease. Neuropsychol. Pharmacol. 39, 2949–2962. doi: 10.1038/npp.2014.164

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, M., Cao, L., and Dai, Y.-P. (2019). Role of neurofilament light chain as a potential biomarker for Alzheimer’s disease: a correlative meta-analysis. Front. Aging Neurosci. 11:254. doi: 10.3389/fnagi.2019.00254

PubMed Abstract | CrossRef Full Text | Google Scholar

Jong, D. D., Kremer, H. P. H., Olde Rikkert, M. G. M., and Verbeek, M. M. (2007). Current state and future directions of neurochemical biomarkers for Alzheimer’s disease. Clin. Chem. Lab. Med. 45, 1421–1434. doi: 10.1515/CCLM.2007.320

PubMed Abstract | CrossRef Full Text | Google Scholar

Jonsson, T., Atwal, J. K., Steinberg, S., Snaedal, J., Jonsson, P. V., Bjornsson, S., et al. (2012). A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature 488, 96–99. doi: 10.1038/nature11283

PubMed Abstract | CrossRef Full Text | Google Scholar

Jonsson, T., Jonsson, P. V., Stefansson, H., Stefansson, K., Steinberg, S., Jonsdottir, I., et al. (2013). Variant of TREM2 associated with the risk of Alzheimer’s disease. New Engl. J. Med. 368, 107–116.

Google Scholar

Kan, C. W., Tobos, C. I., Rissin, D. M., Wiener, A. D., Meyer, R. E., Svancara, D. M., et al. (2020). Digital enzyme-linked immunosorbent assays with sub-attomolar detection limits based on low numbers of capture beads combined with high efficiency bead analysis. Lab Chip 20, 2122–2135. doi: 10.1039/d0lc00267d

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanmert, D., Cantlon, A., Muratore, C. R., Jin, M., O’malley, T. T., Lee, G., et al. (2015). C-terminally truncated forms of tau, but not full-length tau or its C-terminal fragments, are released from neurons independently of cell death. J. Neurosci. 35, 10851–10865. doi: 10.1523/jneurosci.0387-15.2015

PubMed Abstract | CrossRef Full Text | Google Scholar

Karikari, T. K., Benedet, A. L., Ashton, N. J., Lantero Rodriguez, J., Snellman, A., Suárez-Calvet, M., et al. (2020a). Diagnostic performance and prediction of clinical progression of plasma phospho-tau181 in the Alzheimer’s disease neuroimaging initiative. Mol. Psychiatry 26, 429–442. doi: 10.1038/s41380-020-00923-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Karikari, T. K., Pascoal, T. A., Ashton, N. J., Janelidze, S., Benedet, A. L., Rodriguez, J. L., et al. (2020b). 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. 19, 422–433. doi: 10.1016/S1474-4422(20)30071-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Keller, L., Welander, H., Chiang, H.-H., Tjernberg, L. O., Nennesmo, I., Wallin, ÅK., et al. (2010). The PSEN1 I143T mutation in a Swedish family with Alzheimer’s disease: clinical report and quantification of AB in different brain regions. Eur. J. Hum. Genet. 18, 1202–1208. doi: 10.1038/ejhg.2010.107

PubMed Abstract | CrossRef Full Text | Google Scholar

Keshavan, A., Pannee, J., Karikari, T. K., Rodriguez, J. L., Ashton, N. J., Nicholas, J. M., et al. (2021). Population-based blood screening for preclinical Alzheimer’s disease in a British birth cohort at age 70. Brain 144, 434–449. doi: 10.1093/brain/awaa403

PubMed Abstract | CrossRef Full Text | Google Scholar

Kleinberger, G., Yamanishi, Y., Suárez-Calvet, M., Czirr, E., Lohmann, E., Cuyvers, E., et al. (2014). TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci. Transl. Med. 6:243ra286. doi: 10.1126/scitranslmed.3009093

PubMed Abstract | CrossRef Full Text | Google Scholar

Koffie, R. M., Meyer-Luehmann, M., Hashimoto, T., Adams, K. W., Mielke, M. L., Garcia-Alloza, M., et al. (2009). Oligomeric amyloid β associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc. Natl. Acad. Sci. U.S.A. 106, 4012–4017. doi: 10.1073/pnas.0811698106

PubMed Abstract | CrossRef Full Text | Google Scholar

Kohnken, R., Buerger, K., Zinkowski, R., Miller, C., Kerkman, D., Debernardis, J., et al. (2000). Detection of tau phosphorylated at threonine 231 in cerebrospinal fluid of Alzheimer’s disease patients. Neurosci. Lett. 287, 187–190. doi: 10.1016/s0304-3940(00)01178-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Korecka, M., and Shaw, L. M. (2021). Mass spectrometry-based methods for robust measurement of Alzheimer’s disease biomarkers in biological fluids. J. Neurochem. 159, 211–233. doi: 10.1111/jnc.15465

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuchibhotla, K. V., Goldman, S. T., Lattarulo, C. R., Wu, H.-Y., Hyman, B. T., and Bacskai, B. J. (2008). Abeta plaques lead to aberrant regulation of calcium homeostasis in vivo resulting in structural and functional disruption of neuronal networks. Neuron 59, 214–225. doi: 10.1016/j.neuron.2008.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Lauridsen, C., Sando, S. B., Møller, I., Berge, G., Pomary, P. K., Grøntvedt, G. R., et al. (2017). Cerebrospinal fluid Aβ43 is reduced in early-onset compared to late-onset Alzheimer’s disease, but has similar diagnostic accuracy to Aβ42. Front. Aging Neurosci. 9:210. doi: 10.3389/fnagi.2017.00210

PubMed Abstract | CrossRef Full Text | Google Scholar

Lauridsen, C., Sando, S. B., Shabnam, A., Moller, I., Berge, G., Grontvedt, G. R., et al. (2016). Cerebrospinal fluid levels of amyloid beta 1-43 in patients with amnestic mild cognitive impairment or early Alzheimer’s disease: a 2-year follow-up Study. Front. Aging Neurosci. 8:30. doi: 10.3389/fnagi.2016.00030

PubMed Abstract | CrossRef Full Text | Google Scholar

Lewczuk, P., Ermann, N., Andreasson, U., Schultheis, C., Podhorna, J., Spitzer, P., et al. (2018). Plasma neurofilament light as a potential biomarker of neurodegeneration in Alzheimer’s disease. Alzheimers Res. Ther. 10:71. doi: 10.1186/s13195-018-0404-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Lewczuk, P., Esselmann, H., Otto, M., Maler, J. M., Henkel, A. W., Henkel, M. K., et al. (2004). Neurochemical diagnosis of Alzheimer’s dementia by CSF Abeta42. Abeta42/Abeta40 ratio and total tau. Neurobiol. Aging 25, 273–281. doi: 10.1016/S0197-4580(03)00086-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Q. X., Fuller, S. J., Beyreuther, K., and Masters, C. L. (1999). The amyloid precursor protein of Alzheimer disease in human brain and blood. J. Leukocyte Biol. 66, 567–574. doi: 10.1002/jlb.66.4.567

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, Y. S., Lee, W. J., Wang, S. J., and Fuh, J. L. (2018). Levels of plasma neurofilament light chain and cognitive function in patients with Alzheimer or Parkinson disease. Sci. Rep. 8:17368. doi: 10.1038/s41598-018-35766-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Lista, S., Toschi, N., Baldacci, F., Zetterberg, H., Blennow, K., Kilimann, I., et al. (2017). Diagnostic accuracy of CSF neurofilament light chain protein in the biomarker-guided classification system for Alzheimer’s disease. Neurochem. Int. 108, 355–360. doi: 10.1016/j.neuint.2017.05.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Maia, L. F., Kaeser, S. A., Reichwald, J., Hruscha, M., Martus, P., Staufenbiel, M., et al. (2013). Changes in amyloid-β and tau in the cerebrospinal fluid of transgenic mice overexpressing amyloid precursor protein. Sci. Transl. Med. 5:194re192. doi: 10.1126/scitranslmed.3006446

PubMed Abstract | CrossRef Full Text | Google Scholar

Maloney, J. A., Bainbridge, T., Gustafson, A., Zhang, S., Kyauk, R., Steiner, P., et al. (2014). Molecular mechanisms of alzheimer disease protection by the A673T allele of amyloid precursor protein. J. Biol. Chem. 289, 30990–31000. doi: 10.1074/jbc.M114.589069

PubMed Abstract | CrossRef Full Text | Google Scholar

Martin, L., Latypova, X., and Terro, F. (2011). Post-translational modifications of tau protein: implications for Alzheimer’s disease. Neurochem. Int. 58, 458–471. doi: 10.1016/j.neuint.2010.12.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattsson, N., Andreasson, U., Zetterberg, H., Blennow, K., and Neuroimaging, A. S. D. (2017). Association of plasma neurofilament light with neurodegeneration in patients with alzheimer disease. JAMA Neurol. 74, 557–566. doi: 10.1001/jamaneurol.2016.6117

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattsson, N., Insel, P. S., Palmqvist, S., Portelius, E., Zetterberg, H., Weiner, M., et al. (2016). Cerebrospinal fluid tau, neurogranin, and neurofilament light in Alzheimer’s disease. EMBO Mol. Med. 8, 1184–1196. doi: 10.15252/emmm.201606540

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattsson-Carlgren, N., Andersson, E., Janelidze, S., Ossenkoppele, R., Insel, P., Strandberg, O., et al. (2020). Aβ deposition is associated with increases in soluble and phosphorylated tau that precede a positive Tau PET in Alzheimer’s disease. Sci. Adv. 6:eaaz2387. doi: 10.1126/sciadv.aaz2387

PubMed Abstract | CrossRef Full Text | Google Scholar

Mawuenyega, K. G., Sigurdson, W., Ovod, V., Munsell, L., Kasten, T., Morris, J. C., et al. (2010). Decreased clearance of CNS β-amyloid in Alzheimer’s disease. Science 330:1774. doi: 10.1126/science.1197623

PubMed Abstract | CrossRef Full Text | Google Scholar

Mayer, C. A., Brunkhorst, R., Niessner, M., Pfeilschifter, W., Steinmetz, H., and Foerch, C. (2013). Blood levels of glial fibrillary acidic protein (GFAP) in patients with neurological diseases. PLoS One 8:e62101. doi: 10.1371/journal.pone.0062101

PubMed Abstract | CrossRef Full Text | Google Scholar

Metaxas, A., and Kempf, S. J. (2016). Neurofibrillary tangles in Alzheimer’s disease: elucidation of the molecular mechanism by immunohistochemistry and tau protein phospho-proteomics. Neural Regen. Res. 11, 1579–1581. doi: 10.4103/1673-5374.193234

PubMed Abstract | CrossRef Full Text | Google Scholar

Meyer-Luehmann, M., Mielke, M., Spires-Jones, T. L., Stoothoff, W., Jones, P., Bacskai, B. J., et al. (2009). A reporter of local dendritic translocation shows plaque- related loss of neural system function in APP-transgenic mice. J. Neurosci. 29, 12636–12640. doi: 10.1523/JNEUROSCI.1948-09.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Mielke, M. M., Hagen, C. E., Xu, J., Chai, X. Y., Vemuri, P., Lowe, V. J., et al. (2018). Plasma phospho-tau181 increases with Alzheimer’s disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimers Dement. 14, 989–997. doi: 10.1016/j.jalz.2018.02.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Milà-Alomà, M., Salvadó, G., Gispert, J. D., Vilor-Tejedor, N., Grau-Rivera, O., Sala-Vila, A., et al. (2020). Amyloid beta, tau, synaptic, neurodegeneration, and glial biomarkers in the preclinical stage of the Alzheimer’s continuum. Alzheimers Dement. 16, 1358–1371. doi: 10.1002/alz.12131

PubMed Abstract | CrossRef Full Text | Google Scholar

Miller, D. L., Papayannopoulos, I. A., Styles, J., Bobin, S. A., Lin, Y. Y., Biemann, K., et al. (1993). Peptide compositions of the cerebrovascular and senile plaque core amyloid deposits of Alzheimer’s disease. Arch. Biochem. Biophys. 301, 41–52. doi: 10.1006/abbi.1993.1112

PubMed Abstract | CrossRef Full Text | Google Scholar

Molinuevo, J. L., Ayton, S., Batrla, R., Bednar, M. M., Bittner, T., Cummings, J., et al. (2018). Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol. 136, 821–853. doi: 10.1007/s00401-018-1932-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Moore, B. D., Martin, J., De Mena, L., Sanchez, J., Cruz, P. E., Ceballos-Diaz, C., et al. (2018). Short Aβ peptides attenuate Aβ42 toxicity in vivo. J. Exp. Med. 215, 283–301.

Google Scholar

Mori, H., Joachim, C. L., and Selkoe, D. J. (1989). Amyloid β -protein deposition in tissues other than brain in Alzheimer’s disease. Nature 341, 226–230. doi: 10.1038/341226a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Morris, G. P., Clark, I. A., and Vissel, B. (2014). Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer’s disease. Acta Neuropathol. Commun. 2:135. doi: 10.1186/s40478-014-0135-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Nelson, P. T., Alafuzoff, I., Bigio, E. H., Bouras, C., Braak, H., Cairns, N. J., et al. (2012). Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J. Neuropathol. Exp. Neurol. 71, 362–381. doi: 10.1097/NEN.0b013e31825018f7

PubMed Abstract | CrossRef Full Text | Google Scholar

Neumann, H., and Takahashi, K. (2006). Essential role of the microglial triggering receptor expressed on myeloid cells-2 (TREM2) for central nervous tissue immune homeostasis. J. Neuroimmunol. 184, 92–99. doi: 10.1016/j.jneuroim.2006.11.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Oeckl, P., Halbgebauer, S., Anderl-Straub, S., Steinacker, P., Huss, A. M., Neugebauer, H., et al. (2019). Glial fibrillary acidic protein in serum is increased in Alzheimer’s disease and correlates with cognitive impairment. J. Alzheimers Dis. 67, 481–488. doi: 10.3233/JAD-180325

PubMed Abstract | CrossRef Full Text | Google Scholar

Oeckl, P., and Otto, M. (2019). A review on MS-based blood biomarkers for Alzheimer’s disease. Neurol. Ther. 8, 113–127. doi: 10.1007/s40120-019-00165-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohara, T., Hata, J., Tanaka, M., Honda, T., Yamakage, H., Yoshida, D., et al. (2019). Serum soluble triggering receptor expressed on myeloid cells 2 as a biomarker for incident dementia: the hisayama study. Ann. Neurol. 85, 47–58. doi: 10.1002/ana.25385

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsson, B., Lautner, R., Andreasson, U., Ohrfelt, A., Portelius, E., Bjerke, M., et al. (2016). CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 15, 673–684. doi: 10.1016/S1474-4422(16)00070-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Palmqvist, S., Insel, P. S., Stomrud, E., Janelidze, S., Zetterberg, H., Brix, B., et al. (2019). Cerebrospinal fluid and plasma biomarker trajectories with increasing amyloid deposition in Alzheimer’s disease. Embo Mol. Med. 11:e11170. doi: 10.15252/emmm.201911170

PubMed Abstract | CrossRef Full Text | Google Scholar

Palmqvist, S., Janelidze, S., Quiroz, Y. T., Zetterberg, H., Lopera, F., Stomrud, E., et al. (2020). Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. J. Am. Med. Assoc. 324, 772–781. doi: 10.1001/jama.2020.12134

PubMed Abstract | CrossRef Full Text | Google Scholar

Paloneva, J., Autti, T., Raininko, R., Partanen, J., Salonen, O., Puranen, M., et al. (2001). CNS manifestations of Nasu-Hakola disease: a frontal dementia with bone cysts. Neurology 56, 1552–1558. doi: 10.1212/wnl.56.11.1552

PubMed Abstract | CrossRef Full Text | Google Scholar

Paloneva, J., Manninen, T., Christman, G., Hovanes, K., Mandelin, J., Adolfsson, R., et al. (2002). Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am. J. Hum. Genet. 71, 656–662. doi: 10.1086/342259

PubMed Abstract | CrossRef Full Text | Google Scholar

Pannee, J., Törnqvist, U., Westerlund, A., Ingelsson, M., Lannfelt, L., Brinkmalm, G., et al. (2014). The amyloid-β degradation pattern in plasma—A possible tool for clinical trials in Alzheimer’s disease. Neurosci. Lett. 573, 7–12. doi: 10.1016/j.neulet.2014.04.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Paradowska-Gorycka, A., and Jurkowska, M. (2013). Structure, expression pattern and biological activity of molecular complex TREM-2/DAP12. Hum. Immunol. 74, 730–737. doi: 10.1016/j.humimm.2013.02.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Paresce, D. M., Ghosh, R. N., and Maxfield, F. R. (1996). Microglial cells internalize aggregates of the Alzheimer’s disease amyloid β-protein via a scavenger receptor. Neuron 17, 553–565. doi: 10.1016/s0896-6273(00)80187-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, S.-H., Lee, E.-H., Kim, H.-J., Jo, S., Lee, S., Seo, S. W., et al. (2021). The relationship of soluble TREM2 to other biomarkers of sporadic Alzheimer’s disease. Sci. Rep. 11:13050. doi: 10.1038/s41598-021-92101-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Parnetti, L., Lanari, A., Amici, S., Gallai, V., Vanmechelen, E., and Hulstaert, F. (2001). CSF phosphorylated tau is a possible marker for discriminating Alzheimer’s disease from dementia with Lewy bodies. Neurol. Sci. 22, 77–78. doi: 10.1007/s100720170055

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascoal, T. A., Benedet, A. L., Ashton, N. J., Kang, M. S., Therriault, J., Chamoun, M., et al. (2021). Microglial activation and tau propagate jointly across Braak stages. Nat. Med. 27, 1592–1599. doi: 10.1038/s41591-021-01456-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Pera, M., Montesinos, J., Larrea, D., Agrawal, R. R., Velasco, K. R., Stavrovskaya, I. G., et al. (2020). MAM and C99, key players in the pathogenesis of Alzheimer’s disease. Int. Rev. Neurobiol. 154, 235–278. doi: 10.1016/bs.irn.2020.03.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Perea, G., Navarrete, M., and Araque, A. (2009). Tripartite synapses: astrocytes process and control synaptic information. Trends Neurosci. 32, 421–431. doi: 10.1016/j.tins.2009.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Perez-Nievas, B. G., and Serrano-Pozo, A. (2018). Deciphering the astrocyte reaction in Alzheimer’s disease. Front. Aging Neurosci. 10:114. doi: 10.3389/fnagi.2018.00114

PubMed Abstract | CrossRef Full Text | Google Scholar

Perrone, F., Bjerke, M., Hens, E., Sieben, A., Timmers, M., De Roeck, A., et al. (2020). Amyloid-β1–43 cerebrospinal fluid levels and the interpretation of APP, PSEN1 and PSEN2 mutations. Alzheimers Res. Ther. 12:108. doi: 10.1186/s13195-020-00676-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Petzold, A., Keir, G., Warren, J., Fox, N., and Rossor, M. N. (2007). A systematic review and meta-analysis of CSF neurofilament protein levels as biomarkers in dementia. Neurodegener. Dis. 4, 185–194. doi: 10.1159/000101843

PubMed Abstract | CrossRef Full Text | Google Scholar

Piccio, L., Deming, Y., Del-Águila, J. L., Ghezzi, L., Holtzman, D. M., Fagan, A. M., et al. (2016). Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status. Acta Neuropathol. 131, 925–933. doi: 10.1007/s00401-016-1533-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Pike, C. J., Cummings, B. J., and Cotman, C. W. (1995). Early association of reactive astrocytes with senile plaques in Alzheimer’s disease. Exp. Neurol. 132, 172–179. doi: 10.1016/0014-4886(95)90022-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Portelius, E., Bogdanovic, N., Gustavsson, M. K., Volkmann, I., Brinkmalm, G., Zetterberg, H., et al. (2010). Mass spectrometric characterization of brain amyloid beta isoform signatures in familial and sporadic Alzheimer’s disease. Acta Neuropathol. 120, 185–193. doi: 10.1007/s00401-010-0690-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Preische, O., Schultz, S. A., Apel, A., Kuhle, J., Kaeser, S. A., Barro, C., et al. (2019). Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer’s disease. Nat. Med. 25, 277–283. doi: 10.1038/s41591-018-0304-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Pyun, J.-M., Kang, M. J., Ryoo, N., Suh, J., Youn, Y. C., Park, Y. H., et al. (2020). Amyloid Metabolism and Amyloid-Targeting Blood-Based Biomarkers of Alzheimer’s Disease. J. Alzheimers Dis. 75, 685–696. doi: 10.3233/JAD-200104

PubMed Abstract | CrossRef Full Text | Google Scholar

Quiroz, Y. T., Zetterberg, H., Reiman, E. M., Chen, Y., Su, Y., Fox-Fuller, J. T., et al. (2020). Plasma neurofilament light chain in the presenilin 1 E280A autosomal dominant Alzheimer’s disease kindred: a cross-sectional and longitudinal cohort study. Lancet Neurol. 19, 513–521. doi: 10.1016/S1474-4422(20)30137-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Rajan, K. B., Aggarwal, N. T., Mcaninch, E. A., Weuve, J., Barnes, L. L., Wilson, R. S., et al. (2020). Remote blood biomarkers of longitudinal cognitive outcomes in a population study. Ann. Neurol. 88, 1065–1076. doi: 10.1002/ana.25874

PubMed Abstract | CrossRef Full Text | Google Scholar

Ransohoff, R. M., and Perry, V. H. (2009). Microglial physiology: unique stimuli, specialized responses. Annu. Rev. Immunol. 27, 119–145. doi: 10.1146/annurev.immunol.021908.132528

PubMed Abstract | CrossRef Full Text | Google Scholar

Reiman, E. M., Quiroz, Y. T., Fleisher, A. S., Chen, K., Velez-Pardo, C., Jimenez-Del-Rio, M., et al. (2012). Brain imaging and fluid biomarker analysis in young adults at genetic risk for autosomal dominant Alzheimer’s disease in the presenilin 1 E280A kindred: a case-control study. Lancet Neurol. 11, 1048–1056. doi: 10.1016/S1474-4422(12)70228-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Riemenschneider, M., Wagenpfeil, S., Vanderstichele, H., Otto, M., Wiltfang, J., Kretzschmar, H., et al. (2003). Phospho-tau total tau ratio in cerebrospinal fluid discriminates Creutzfeldt-Jakob disease from other dementias. Mol. Psychiatry 8, 343–347. doi: 10.1038/sj.mp.4001220

PubMed Abstract | CrossRef Full Text | Google Scholar

Rissin, D. M., Kan, C. W., Campbell, T. G., Howes, S. C., Fournier, D. R., Song, L., et al. (2010). Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat. Biotechnol. 28, 595–599. doi: 10.1038/nbt.1641

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez-Vieitez, E., and Ashton, N. J. (2021). Plasma sTREM2: a potential marker of cerebrovascular injury in neurodegenerative disorders. Brain 144, 3283–3285. doi: 10.1093/brain/awab399

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez-Vieitez, E., Saint-Aubert, L., Carter, S. F., Almkvist, O., Farid, K., Schöll, M., et al. (2016). Diverging longitudinal changes in astrocytosis and amyloid PET in autosomal dominant Alzheimer’s disease. Brain 139, 922–936. doi: 10.1093/brain/awv404

PubMed Abstract | CrossRef Full Text | Google Scholar

Roher, A. E., Esh, C. L., Kokjohn, T. A., Castaño, E. M., Van Vickle, G. D., Kalback, W. M., et al. (2009). Aβ peptides in human plasma and tissues and their significance for Alzheimer’s disease. Alzheimers Dement. 5, 18–29. doi: 10.1016/j.jalz.2008.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Rohrer, J. D., Woollacott, I. O. C., Dick, K. M., Brotherhood, E., Gordon, E., Fellows, A., et al. (2016). Serum neurofilament light chain protein is a measure of disease intensity in frontotemporal dementia. Neurology 87, 1329–1336. doi: 10.1212/WNL.0000000000003154

PubMed Abstract | CrossRef Full Text | Google Scholar

Rojas, J. C., Karydas, A., Bang, J., Tsai, R. M., Blennow, K., Liman, V., et al. (2016). Plasma neurofilament light chain predicts progression in progressive supranuclear palsy. Ann. Clin. Transl. Neurol. 3, 216–225. doi: 10.1002/acn3.290

PubMed Abstract | CrossRef Full Text | Google Scholar

Rosenthal, S. L., Bamne, M. N., Wang, X., Berman, S., Snitz, B. E., Klunk, W. E., et al. (2015). More evidence for association of a rare TREM2 mutation (R47H) with Alzheimer’s disease risk. Neurobiol. Aging 36:2443. doi: 10.1016/j.neurobiolaging.2015.04.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Saito, T., Suemoto, T., Brouwers, N., Sleegers, K., Funamoto, S., Mihira, N., et al. (2011). Potent amyloidogenicity and pathogenicity of Aβ43. Nat. Neurosci. 14, 1023–1032. doi: 10.1038/nn.2858

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez-Valle, R., Heslegrave, A., Foiani, M. S., Bosch, B., Antonell, A., Balasa, M., et al. (2018). Serum neurofilament light levels correlate with severity measures and neurodegeneration markers in autosomal dominant Alzheimer’s disease. Alzheimers Res. Ther. 10:113. doi: 10.1186/s13195-018-0439-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez-Varo, R., Trujillo-Estrada, L., Sanchez-Mejias, E., Torres, M., Baglietto-Vargas, D., Moreno-Gonzalez, I., et al. (2011). Abnormal accumulation of autophagic vesicles correlates with axonal and synaptic pathology in young Alzheimer’s mice hippocampus. Acta Neuropathol. 123, 53–70. doi: 10.1007/s00401-011-0896-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sato, C., Barthélemy, N. R., Mawuenyega, K. G., Patterson, B. W., Gordon, B. A., Jockel-Balsarotti, J., et al. (2018). Tau kinetics in neurons and the human central nervous system. Neuron 98, 861–864.

Google Scholar

Schilling, S., Lauber, T., Schaupp, M., Manhart, S., Scheel, E., Böhm, G., et al. (2006). On the seeding and oligomerization of pGlu-amyloid peptides (in vitro). Biochemistry 45, 12393–12399. doi: 10.1021/bi0612667

PubMed Abstract | CrossRef Full Text | Google Scholar

Schlaepfer, W. W., and Lynch, R. G. (1977). Immunofluorescence studies of neurofilaments in the rat and human peripheral and central nervous system. J. Cell Biol. 74, 241–250. doi: 10.1083/jcb.74.1.241

PubMed Abstract | CrossRef Full Text | Google Scholar

Scholl, M., Carter, S. F., Westman, E., Rodriguez-Vieitez, E., Almkvist, O., Thordardottir, S., et al. (2015). Early astrocytosis in autosomal dominant Alzheimer’s disease measured in vivo by multi-tracer positron emission tomography. Sci. Rep. 5:16404. doi: 10.1038/srep16404

PubMed Abstract | CrossRef Full Text | Google Scholar

Shekhar, S., Kumar, R., Rai, N., Kumar, V., Singh, K., Upadhyay, A. D., et al. (2016). Estimation of Tau and phosphorylated Tau181 in serum of Alzheimer’s disease and mild cognitive impairment patients. PLoS One 11:e0159099. doi: 10.1371/journal.pone.0159099

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, L., Baird, A. L., Westwood, S., Hye, A., Dobson, R., Thambisetty, M., et al. (2018). A decade of blood biomarkers for Alzheimer’s disease research: an evolving field, improving study designs, and the challenge of replication. J. Alzheimers Dis. 62, 1181–1198. doi: 10.3233/JAD-170531

PubMed Abstract | CrossRef Full Text | Google Scholar

Sjögren, M., Blomberg, M., Jonsson, M., Wahlund, L. O., Edman, Å, Lind, K., et al. (2001). Neurofilament protein in cerebrospinal fluid: a marker of white matter changes. J. Neurosci. Res. 66, 510–516. doi: 10.1002/jnr.1242

PubMed Abstract | CrossRef Full Text | Google Scholar

Slattery, C. F., Beck, J. A., Harper, L., Adamson, G., Abdi, Z., Uphill, J., et al. (2014). R47H TREM2 variant increases risk of typical early-onset Alzheimer’s disease but not of prion or frontotemporal dementia. Alzheimers Dement. 10:602. doi: 10.1016/j.jalz.2014.05.1751

PubMed Abstract | CrossRef Full Text | Google Scholar

Smith, R. W. (2013). Mass Spectrometry. Amsterdam: Elsevier, 603–608.

Google Scholar

Söllvander, S., Nikitidou, E., Brolin, R., Söderberg, L., Sehlin, D., Lannfelt, L., et al. (2016). Accumulation of amyloid-β by astrocytes result in enlarged endosomes and microvesicle-induced apoptosis of neurons. Mol. Neurodegener. 11:38. doi: 10.1186/s13024-016-0098-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, L., Zhao, M., Duffy, D. C., Hansen, J., Shields, K., Wungjiranirun, M., et al. (2015). Development and validation of digital enzyme-linked immunosorbent assays for ultrasensitive detection and quantification of Clostridium difficile toxins in stool. J. Clin. Microbiol. 53, 3204–3212. doi: 10.1128/JCM.01334-15

PubMed Abstract | CrossRef Full Text | Google Scholar

Stelzmann, R. A., Norman Schnitzlein, H., and Reed Murtagh, F. (1995). An english translation of alzheimer’s 1907 paper, “über eine eigenartige erkankung der hirnrinde”. Clin. Anat. 8, 429–431. doi: 10.1002/ca.980080612

PubMed Abstract | CrossRef Full Text | Google Scholar

Struyfs, H., Van Broeck, B., Timmers, M., Fransen, E., Sleegers, K., Van Broeckhoven, C., et al. (2015). Diagnostic accuracy of cerebrospinal fluid amyloid-beta isoforms for early and differential dementia diagnosis. J. Alzheimers Dis. 45, 813–822. doi: 10.3233/JAD-141986

PubMed Abstract | CrossRef Full Text | Google Scholar

Suárez-Calvet, M., Caballero, M. ÁA., Kleinberger, G., Bateman, R. J., Fagan, A. M., Morris, J. C., et al. (2016). Early changes in CSF sTREM2 in dominantly inherited Alzheimer’s disease occur after amyloid deposition and neuronal injury. Sci. Transl. Med. 8:369ra178. doi: 10.1126/scitranslmed.aag1767

PubMed Abstract | CrossRef Full Text | Google Scholar

Suarez-Calvet, M., Kleinberger, G., Caballero, M. A. A., Brendel, M., Rominger, A., Alcolea, D., et al. (2016). 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. 8, 466–476. doi: 10.15252/emmm.201506123

PubMed Abstract | CrossRef Full Text | Google Scholar

Suárez-Calvet, M., Karikari, T. K., Ashton, N. J., Lantero Rodríguez, J., Milà-Alomà, M., Gispert, J. D., et al. (2020). Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer’s continuum when only subtle changes in Aβ pathology are detected. EMBO Mol. Med. 12:e12921.

Google Scholar

Subramanian, J., Savage, J. C., and Tremblay, M. -È (2020). Synaptic Loss in Alzheimer’s disease: mechanistic insights provided by two-photon in vivo imaging of transgenic mouse models. Front. Cell. Neurosci. 14:592607. doi: 10.3389/fncel.2020.592607

PubMed Abstract | CrossRef Full Text | Google Scholar

Tatebe, H., Kasai, T., Ohmichi, T., Kishi, Y., Kakeya, T., Waragai, M., et al. (2017). 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. 12:63. doi: 10.1186/s13024-017-0206-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Thal, D. R., Schultz, C., Dehghani, F., Yamaguchi, H., Braak, H., and Braak, E. (2000). Amyloid beta-protein (Abeta)-containing astrocytes are located preferentially near N-terminal-truncated Abeta deposits in the human entorhinal cortex. Acta Neuropathol. 100, 608–617. doi: 10.1007/s004010000242

PubMed Abstract | CrossRef Full Text | Google Scholar

Ulrich, J. D., Ulland, T. K., Colonna, M., and Holtzman, D. M. (2017). Elucidating the role of TREM2 in Alzheimer’s disease. Neuron 94, 237–248. doi: 10.1016/j.neuron.2017.02.042

PubMed Abstract | CrossRef Full Text | Google Scholar

van Ballegoij, W. J. C., Van De Stadt, S. I. W., Huffnagel, I. C., Kemp, S., Willemse, E. A. J., Teunissen, C. E., et al. (2020). Plasma NfL and GFAP as biomarkers of spinal cord degeneration in adrenoleukodystrophy. Ann. Clin. Transl. Neurol. 7, 2127–2136. doi: 10.1002/acn3.51188

PubMed Abstract | CrossRef Full Text | Google Scholar

Vanmechelen, E., Vanderstichele, H., Davidsson, P., Van Kerschaver, E., Van Der Perre, B., Sjögren, M., et al. (2000). Quantification of tau phosphorylated at threonine 181 in human cerebrospinal fluid: a sandwich ELISA with a synthetic phosphopeptide for standardization. Neurosci. Lett. 285, 49–52. doi: 10.1016/s0304-3940(00)01036-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Vehmas, A. K., Kawas, C. H., Stewart, W. F., and Troncoso, J. C. (2003). Immune reactive cells in senile plaques and cognitive decline in Alzheimer’s disease. Neurobiol. Aging 24, 321–331. doi: 10.1016/s0197-4580(02)00090-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Verbeek, M. M., Kremer, B. P., Jansen, R. W., and De Jong, D. (2004). Tau protein phosphorylated at threonine 181 in cerebrospinal fluid as a possible biomarker for Alzheimer’s disease. Neurobiol. Aging 25, S364–S364.

Google Scholar

Verberk, I. M. W., Thijssen, E., Koelewijn, J., Mauroo, K., Vanbrabant, J., De Wilde, A., et al. (2020). Combination of plasma amyloid beta(1-42/1-40) and glial fibrillary acidic protein strongly associates with cerebral amyloid pathology. Alzheimers Res. Ther. 12, 1–118. doi: 10.1186/s13195-020-00682-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Vergallo, A., Megret, L., Lista, S., Cavedo, E., Zetterberg, H., Blennow, K., et al. (2019). Plasma amyloid beta 40/42 ratio predicts cerebral amyloidosis in cognitively normal individuals at risk for Alzheimer’s disease. Alzheimers Dement. 15, 764–775. doi: 10.1016/j.jalz.2019.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Villemagne, V. L., Burnham, S., Bourgeat, P., Brown, B., Ellis, K. A., Salvado, O., et al. (2013). Amyloid β deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: a prospective cohort study. Lancet Neurol. 12, 357–367. doi: 10.1016/S1474-4422(13)70044-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Villemagne, V. L., Doré, V., Burnham, S. C., Masters, C. L., and Rowe, C. C. (2018). Imaging tau and amyloid-? proteinopathies in Alzheimer disease and other conditions. Nat. Rev. Neurol. 14, 225–236. doi: 10.1038/nrneurol.2018.9

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Cella, M., Mallinson, K., Ulrich, J. D., Young, K. L., Robinette, M. L., et al. (2015). TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell 160, 1061–1071. doi: 10.1016/j.cell.2015.01.049

PubMed Abstract | CrossRef Full Text | Google Scholar

Welander, H., Frånberg, J., Graff, C., Sundström, E., Winblad, B., and Tjernberg, L. O. (2009). Aβ43 is more frequent than Aβ40 in amyloid plaque cores from Alzheimer disease brains. J. Neurochem. 110, 697–706. doi: 10.1111/j.1471-4159.2009.06170.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Welge, V., Fiege, O., Lewczuk, P., Mollenhauer, B., Esselmann, H., Klafki, H. W., et al. (2009). Combined CSF tau, p-tau181 and amyloid-beta 38/40/42 for diagnosing Alzheimer’s disease. J. Neural Trans. 116, 203–212. doi: 10.1007/s00702-008-0177-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Weston, P. S. J., Poole, T., O’connor, A., Heslegrave, A., Ryan, N. S., Liang, Y., et al. (2019). Longitudinal measurement of serum neurofilament light in presymptomatic familial Alzheimer’s disease. Alzheimers Res. Ther. 11:19. doi: 10.1186/s13195-019-0472-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Weydt, P., Oeckl, P., Huss, A., Müller, K., Volk, A. E., Kuhle, J., et al. (2016). Neurofilament levels as biomarkers in asymptomatic and symptomatic familial amyotrophic lateral sclerosis. Ann. Neurol. 79, 152–158. doi: 10.1002/ana.24552

PubMed Abstract | CrossRef Full Text | Google Scholar

Winner, B., Kohl, Z., and Gage, F. H. (2011). Neurodegenerative disease and adult neurogenesis. Eur. J. Neurosci. 33, 1139–1151. doi: 10.1111/j.1460-9568.2011.07613.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wiseman, F. K., Al-Janabi, T., Hardy, J., Karmiloff-Smith, A., Nizetic, D., Tybulewicz, V. L. J., et al. (2015). A genetic cause of Alzheimer disease: mechanistic insights from Down syndrome. Nat. Rev. Neurosci. 16, 564–574. doi: 10.1038/nrn3983

PubMed Abstract | CrossRef Full Text | Google Scholar

Wyss-Coray, T., Yan, F., Brionne, T. C., Lu, E., Husemann, J., Loike, J. D., et al. (2003). Adult mouse astrocytes degrade amyloid-β in vitro and in situ. Nat. Med. 9, 453–457. doi: 10.1038/nm838

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, A., and Nixon, R. A. (2021). Neurofilament proteins as biomarkers to monitor neurological diseases and the efficacy of therapies. Front. Neurosci. 15:689938. doi: 10.3389/fnins.2021.689938

PubMed Abstract | CrossRef Full Text | Google Scholar

Zetterberg, H., and Burnham, S. C. (2019). Blood-based molecular biomarkers for Alzheimer’s disease. Mol. Brain 12:26.

Google Scholar

Zetterberg, H., Skillbäck, T., Mattsson, N., Trojanowski, J. Q., Portelius, E., Shaw, L. M., et al. (2016). Association of cerebrospinal fluid neurofilament light concentration with Alzheimer disease progression. JAMA Neurol. 73, 60–67. doi: 10.1001/jamaneurol.2015.3037

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: amyloid-beta, blood biomarkers, neurodegeneration, neurofilament light (NfL), glial fibrillary acidic protein (GFAP), phosphorylated tau (p-tau), triggering receptor expressed on myeloid cells 2 (TREM2)

Citation: Alawode DOT, Fox NC, Zetterberg H and Heslegrave AJ (2022) Alzheimer’s Disease Biomarkers Revisited From the Amyloid Cascade Hypothesis Standpoint. Front. Neurosci. 16:837390. doi: 10.3389/fnins.2022.837390

Received: 16 December 2021; Accepted: 04 April 2022;
Published: 27 April 2022.

Edited by:

Can Martin Zhang, Massachusetts General Hospital and Harvard Medical School, United States

Reviewed by:

Christian Humpel, Innsbruck Medical University, Austria
Cristina Lanni, University of Pavia, Italy
Agata Copani, University of Catania, Italy
Masato Maesako, Massachusetts General Hospital and Harvard Medical School, United States

Copyright © 2022 Alawode, Fox, Zetterberg and Heslegrave. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Deborah O. T. Alawode, d.alawode.16@ucl.ac.uk; Amanda J. Heslegrave, a.heslegrave@ucl.ac.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.