Next Article in Journal
The Multifaceted Roles of Ku70/80
Next Article in Special Issue
Understanding Amyloid Structures and Disease: A Continuing Challenge in Health Research
Previous Article in Journal
Emerging Roles of Gut Virome in Pediatric Diseases
Previous Article in Special Issue
Diphenyl-Methane Based Thyromimetic Inhibitors for Transthyretin Amyloidosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Diagnostic Potential of Amyloidogenic Proteins

Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(8), 4128; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084128
Submission received: 24 March 2021 / Revised: 12 April 2021 / Accepted: 13 April 2021 / Published: 16 April 2021

Abstract

:
Neurodegenerative disorders are a highly prevalent class of diseases, whose pathological mechanisms start before the appearance of any clear symptoms. This fact has prompted scientists to search for biomarkers that could aid early treatment. These currently incurable pathologies share the presence of aberrant aggregates called amyloids in the nervous system, which are composed of specific proteins. In this review, we discuss how these proteins, their conformations and modifications could be exploited as biomarkers for diagnostic purposes. We focus on proteins that are associated with the most prevalent neurodegenerative disorders, including Alzheimer’s and Parkinson’s diseases, amyotrophic lateral sclerosis, and frontotemporal dementia. We also describe current challenges in detection, the most recent techniques with diagnostic potentials and possible future developments in diagnosis.

1. The Need for Novel Diagnostic Approaches for Neurodegeneration

Neurodegenerative diseases are fatal and incurable disorders, characterized by the progressive loss of neurons in specific regions of the nervous system. They are a highly heterogeneous group of pathologies, which include Alzheimer’s disease (AD), Parkinson’s disease (PD), frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Currently, worldwide, more than 50 million people suffer from various forms of neurodegeneration [1].
The clinical course of neurodegenerative diseases usually spans several years and leads to progressive deficits in memory, cognition, and movement to different extents depending on the specific pathology [2]. Existing drug treatments focus on the relief of these symptoms [3]. Furthermore, neuropsychological assessment is still considered crucial in the diagnosis neurodegeneration associated with dementia [4]. However, it is apparent that key molecular mechanisms of disease occur before the appearance of any significant symptoms. The development of accurate diagnostic approaches would facilitate timely therapeutic interventions to restore neuronal physiology before irreversible damage occurs. It would also promote the establishment of new therapeutics, and the revaluation of current ones which could be more effective if administrated at earlier stages. Despite the urgent need for diagnostic approaches for neurodegeneration, their development is still a daunting challenge, due to the limited accessibility of the brain for physical examination and the complexity of clinical tests based on cognitive abilities [2].
Recent technological advances have enabled the characterization of novel pathways, biomolecules, and structures in the nervous system and other regions of the body that could be used as disease markers of neurodegeneration [5,6]. Although the different neurodegenerative diseases have some distinct phenotypes, they also share some key molecular features. In particular, in many of these disorders, specific proteins and peptides, which would normally be soluble, undergo a self-assembly process which leads to the formation of large fibrillar aggregates, called amyloids [7,8]. This process also involves the generation of smaller oligomeric intermediates, which are highly toxic and currently regarded as major players in the disease mechanisms [8]. Amyloid aggregation is also linked to other aggregation processes, such as the formation of condensates [9]. However, these will not be the focus of the present review.
Here, we describe some of the most known amyloidogenic proteins and their diagnostic relevance. We focus on amyloid-beta (Aβ) and tau for AD, α-synuclein (α-syn) for PD, fused in sarcoma (FUS) and the TAR DNA-binding protein 43 (TDP-43) for ALS and FTD. We also discuss state-of-the-art advancements in detection approaches to monitor the aggregation of these proteins.

2. Amyloid Aggregation as Potential Source of Biomarkers

Amyloids are insoluble fibrillar aggregates enriched in a cross-β structure, and their formation has been extensively characterized in vitro [7,8]. Amyloid aggregation consists of a complex network of nucleation events. Initially, soluble monomeric proteins interact and form oligomers by primary nucleation. Primary nucleation can also be triggered by the presence of other biomolecules, such as other proteins [10], nucleic acids [11,12] and membranes [13]. Oligomers then convert into higher-order aggregates and, finally, into amyloid fibrils. Once a critical concentration of fibrils has formed, the surface of these fibrils catalyzes the formation of additional oligomers by secondary nucleation [7,8]. Fibrils can also elongate by the addition of monomers at their ends, and undergo fragmentation [7,8].
Amyloid fibrils are extremely stable protein species due to their rich cross β-sheet content [8,14,15,16]. On the contrary, oligomers rapidly convert into higher order amyloid aggregates (Figure 1). The transient nature of oligomers makes them difficult to isolate and characterize at the structural level. Only recently, thanks to new approaches such as single molecule fluorescence and electron microscopy [14,17,18], has it been shown that oligomers are highly heterogeneous in their physiochemical properties and structures, with a varying secondary structure content [16,19,20].
Oligomers are toxic by a multitude of mechanisms including aberrant hydrophobic interactions. It has been shown that, in tissue and isolated cells, oligomers can affect membrane permeability, ion homeostasis, and induce oxidative stress [16,21,22,23]. Oligomer-induced free radicals can then trigger protein misfolding, mitochondrial dysfunction and eventually apoptosis [24]. Oligomers of Aβ and α-syn have also been associated with neuroinflammation [22] and synapsis loss [25,26].
Several factors can affect the formation of amyloid fibrils and oligomers. These include genetic mutations, cellular stress, and the presence or absence of specific biomolecules. Furthermore, amyloids are extensively post-translationally modified in vivo [27,28,29] and post-translational modifications (PTMs) significantly alter the formation and the toxicity of amyloid fibrils in vitro [27,28,29,30,31].

3. Amyloidogenic Proteins Involved in Neurodegeneration

In the context of neurodegeneration, disease markers can be divided into neuropsychological, neuroimaging, genetic and biochemical markers [2]. In particular, biochemical markers (or biomarkers) are measurable molecules in our body (e.g., proteins, nucleic acids, metabolites), which report the stage of a disease [32]. Amyloidogenic proteins are promising biomarkers, as they inform on the biochemical profile of the nervous system dysfunction [2]. Below, we highlight relevant amyloidogenic proteins and their pathological modifications which can serve as biomarkers for neurodegenerative conditions.

3.1. Aβ and Tau in AD

AD is the most prevalent form of dementia. The characteristic lesions in AD brains are extracellular senile plaques composed of amyloid aggregates of Aβ and intracellular neurofibrillary tangles (NFTs) formed by paired helical amyloid filaments (PHFs) of hyperphosphorylated tau (p-tau) protein [2,27].
Aβ is a short peptide generated by the cleavage of a larger transmembrane precursor, called the amyloid precursor protein (APP), by the sequential cleavage of the β- and γ-secretases and released into the extracellular space [2,33]. This process can generate Aβ isoforms of various lengths (Table 1), which have various degrees of toxicity in the context of AD [34]. The most common Aβ isoforms are the 40- and 42-residue long ones, generally referred to as Aβ40 and Aβ42, respectively. Aβ40 is the most abundant variant in the plaques (~80% to 90%) and is also present in healthy peoples’ brains. Aβ42 has a much higher propensity to aggregate, and an increase in Aβ42/Aβ40 ratios is associated with AD and other forms of dementia [30,34] (Table 2). Besides cleavage, genetic mutations (A692G, E693Q in the APP gene [35,36], Table 2) and many other PTMs of Aβ have been associated with AD, including oxidation, phosphorylation, glycosylation and isomerization [30]. Studies show the occurrence of acetylation (e.g., Lys16 and Lys28), phosphorylation (e.g., Ser8 and Ser26), nitration (e.g., Tyr10), pyroglutamation (e.g., Glu3 and Glu11), isomerization (e.g., Asp1 and Asp7) and racemization (e.g., Asp1, Asp23 and Ser26) in the context of disease [30,37,38,39] (Table 1). It is worth noting that cognitive decline correlates more with soluble intermediate forms of Aβ rather than the degree of amyloid deposits [16].
Tau is a major microtubule-associated protein that stabilizes the microtubules in neurons [2,40]. In human brains, tau exists as six different isoforms that carry either three or four microtubule-binding repeats (R). These isoforms are called 3R and 4R, respectively. It has been found that the presence of either 3R or 4R or both 3R and 4R amyloids is disease specific. As an example, in AD, ALS, FTD and Parkinsonism, both 3R and 4R amyloids are present, while in corticobasal degenerations and Pick’s disease only 4R and 3R amyloids, respectively, are found [41,42]. Tau undergoes PTMs, particularly phosphorylation [40]. Pathological hyperphosphorylation reduces tau affinity for microtubules and causes its detachment from microtubules, resulting in the formation of PHFs and NFTs [2]. To date, 85 potential phosphorylation sites of tau have been identified [40]. Furthermore, molecular and cellular studies revealed that acetylation (e.g., Lys174, Lys274 and Lys280), oxidation (e.g., Cys322), nitration (e.g., Tyr29), glycation (e.g., Lys87, Lys132 and Lys150), truncation (e.g., at Asp13 and Asp421 and Glu391) and ubiquitination (e.g., Lys48 and Lys63) also affect tau aggregation [27] (Table 1).

3.2. α-Syn in PD

Unlike AD, PD primarily affects the motor system, causing tremors, rigidity, bradykinesia and postural instability [2]. The pathological hallmark of PD is the occurrence of cytoplasmic amyloid inclusions, known as Lewy bodies (LBs) and Lewy neurites (LNs). LBs and LNs are comprised of amyloid aggregates, whose main component is α-syn [43].
α-Syn contains 140 residues with a positively charged N-terminal region, an aggregation-prone non-amyloid-β component (NAC) central region and a negatively charged C-terminal region [44,45]. Duplications or triplications on the α-syn chromosome region (4q21-23) and mutations including A53T, G51D, H50Q, E46K, and A30P in the α-syn sequence, are associated with early-onset PD [46,47,48] (Table 2).
α-Syn belongs to a protein family which also includes β- and γ-synucleins with 55%–62% similarity. β-Synuclein has a reduced propensity to aggregate and has been discovered to suppress the aggregation of α-syn as a natural inhibitor while oxidized γ-synuclein can initiate α-syn aggregation [49,50]. Several PTMs are known to affect aggregation of α-syn, are associated with PD [28] and hold diagnostic potential [51] (Table 1). These include N-terminal acetylation, several truncations at the N-terminus (e.g., α-syn7-140, 14-140, 40-140, and 72–140 found in vitro, 5–140 and 68–140 found in vivo and several in both) and C-terminus (e.g., α-syn1-115, 1-119, 1-122, 1-124, 1-125, 1-129, 1-133, and 1-135), phosphorylation of Ser87 and Ser129, oxidation of Met1, Met5, Met116 and Met127, sumolyation of Lys96 and Lys102, nitration of Tyr39, Tyr125 and Tyr133, and ubiquitination of Lys6, Lys10, Lys12 Lys21, Lys23, Lys43 and Lys96 [28,52,53] (Table 1).

3.3. TDP-43 and FUS in ALS and FTD

ALS and FTD are neurodegenerative diseases with overlapping mechanisms. ALS affects upper and lower neurons, causing loss of muscle control. FTD is a form of dementia linked to the degeneration of the frontal and anterior temporal lobes [54]. Around 97% of ALS and 45% of FTD cases are associated with the presence of inclusions of aggregates of ubiquinated, hyperphosphorylated and C-terminally truncated TDP-43 in the cytoplasm of neurons and glial cells [29].
TDP-43 is a 414 residue-long ribonucleoprotein able to form amyloid-like aggregates in vitro and condensates (i.e., stress granules) [9] under pathological conditions. It is composed of an N-terminal tract with a nuclear localization signal, two RNA recognition motifs, a nuclear export signal, and a disordered C-terminal region [29]. All these regions have been reported to play a critical role in the aggregation of the protein [55,56,57]. Several TDP-43 mutations have been identified in both sporadic and familial cases of ALS and FTD, including G294A, Q331K, M337V [58] and K181E [59] (Table 2). As with Aβ and α-syn, PTMs of TDP-43 also play a key role in the aggregation of the protein and disease progression. It is worth noting that the truncated 25 kDa and 35 kDa C-terminal fragments are commonly found in pathological aggregates in ALS patients [29,31,54]. Ubiquitination is also a typical modification of TDP-43 inclusions [31]. Finally, aberrant phosphorylation, acetylation, and oxidation of TDP-43 is often associated with the mislocalization and aberrant aggregation of the protein [29] (Table 1).
ALS and FTD are also associated with another RNA/DNA-binding protein, FUS. FUS is a 526 residue-long protein made by an N-terminal transcriptional activation domain and C-terminal domain, which interacts with transcriptional factors and also includes a nuclear localization signal [60,61]. Both domains contain low complexity regions and play a role in the formation of condensates and hydrogels [62]. More than 50 mutations (e.g., R521C, R521H [63]) of FUS are reported in ALS/FTD cases. Unlike TDP-43, FUS is generally found as a full-length protein in the aggregates [61]. Phosphorylation occurs in the prion-like domains of FUS and has been shown to affect its phase separation and aggregation pattern while mutations and PTMs (mainly methylation and phosphorylation) on its C-terminal domain are found to regulate its nuclear/cytoplasm localization [61,64]. Despite the clear pathological role of FUS, ALS/FTD phenotypes are less frequently associated with FUS than with TDP-43 dysfunctions [61,65]. Thus, the role of FUS as biomarker remains to be determined.

4. Diagnostic Potential of Genetic, Structural and Chemical Features of Amyloidogenic Proteins

Several detection approaches have been used to quantify amyloidogenic proteins in biological samples and to determine the link between these proteins and neurodegenerative diseases. Some of these strategies aim at quantifying the changes in the expression levels/concentration of amyloidogenic proteins regardless of their conformation or modification (Table 2). Some other approaches, instead, focus on probing specific structural (e.g., aggregated states) or chemical (i.e., PTM) properties of amyloidogenic proteins (Table 1).
All these approaches analyze different regions of the body. Techniques, such as positron emission tomography (PET), are able to probe proteins directly within the central nervous system (CNS), for example, in the brain [2,44,66]. However, amyloidogenic proteins can be also detected in other more accessible regions of the body. In this section, we discuss findings which have been obtained from the analysis of both brain tissues and accessible body fluids, mainly by means of immunoassays, such as immunoblotting and enzyme-linked immunosorbent assays (ELISA). The fluids under consideration include the cerebrospinal fluid (CSF), which is in direct contact with the extracellular portion of the brain and, as such, is an optimal fluid for measurements of brain metabolism [44].

4.1. Levels of Amyloidogenic Proteins in Neurodegeneration

So far, the most common detection strategies probe specific amyloidogenic proteins, regardless of their conformations or PTMs, providing information on the total amount of amyloidogenic protein in disease conditions. These methods are particularly effective when the pathological mechanism depends on genetic mutations (e.g., when a genetic variant of a protein is specifically expressed in patients, Table 2), protein expression levels (e.g., when protein expression is significantly altered in patients) or mislocalization (e.g., when a protein accumulates in specific regions of the body in patients).
In the case of AD, initial experiments focused on measuring the CSF concentration of total Aβ. However, no significant differences between patients and controls were found [33]. In contrast, significant differences have been observed for total tau. Researchers found a marked increase of total tau in the CSF of AD patients compared to healthy controls [67]. Although there is a clear link between the concentration of tau in the CSF and AD, higher concentrations of tau are also observed in acute stroke, brain trauma and other forms of dementia [66,68], making disease-specific diagnosis challenging [44,66].
In the case of PD, it has been found that α-syn is present at slightly lower concentrations in the CSF of patients compared to healthy individuals in a small cohort studies [68,69,70]. This has been observed also in plasma and saliva. In larger studies, CSF α-syn has a significantly lower concentration than healthy controls [69]. This lower concentration of α-syn in accessible body fluids may be a consequence of the intracellular aggregation of the protein [69]. However, the use of α-syn alone is not sufficient as a single biomarker for PD diagnosis [69].
Due to its low concentration, TDP-43 is challenging to reliably measure in the blood [71,72]. However, in the CSF, it has been shown that TDP-43 concentration is higher in ALS patients (6.92 ± 3.71 ng/mL) than in healthy subjects (5.31 ± 0.94 ng/mL) and people affected by other conditions, for example, PD, multiple sclerosis, and Guillain-Barré syndrome [73]. Recent studies have suggested that TDP-43 present in the CSF could be a biomarker for the diagnosis of ALS, ALS/FTD but not for FTD alone [74,75]. In particular, it should be noted that proteins, including TDP-43, can diffuse from the blood into the CSF. Furthermore, TDP-43 is also abundant in other parts of the body besides the brain [54,76]. Thus, TDP-43 in the CSF may originate from other body regions and may not adequately represent the brain pathology in FTD. Currently, research efforts are focusing on monitoring TDP-43 contained in CSF exosomes, which are brain-derived and thus better represent the brain pathology [77]. Overall, so far, despite compelling evidence of the fact that protein expression and concentrations are altered in disease, probing total protein concentration alone has not yielded a widely adopted diagnostic approach.
Table 1. Main PTMs of amyloidogenic proteins with diagnostic potential.
Table 1. Main PTMs of amyloidogenic proteins with diagnostic potential.
ProteinPTMsMajor Modification SitesKey RemarksRefs.
Cleavage1-37, 1-38, 1-39, 1-40, 1-42, 1-43Aβ 40 is the most abundant; Aβ 42 aggregates more rapidly.[30,78,79]
PhosphorylationSer8, Ser26Increased abundance and stability of toxic aggregates.[30,80,81]
AcetylationLys16, Lys28Altered aggregation behavior.[38,39]
OxidationMet35Regulation of oxidative stress. Slower aggregation.[30,82]
NitrationTyr10Enhanced aggregation and plaque formation.[30,37]
IsomerizationAsp1, Asp7Higher aggregation propensity and resistance to degradation.[30]
RacemizationAsp1, Asp23, Ser26Higher aggregation propensity.[30]
O-glycosylationTyr10Found in short Aβ fragments in AD patients’ CSF. Aβ1-15 and Aβ1-17 are the most abundant fragments.[30,83]
Pyroglutamate formationGlu3, Glu11Increased oligomerization. It correlates with the extent of Aβ deposition.[84,85]
TauPhosphorylationThr181, Thr199, Thr217, Thr231Increased aggregation. It is a key event in the formation of NFT.[27,86]
AcetylationLys174, Lys274, Lys280Regulation of tau function; promotion of p-tau aggregation.[87]
OxidationCys322Enhanced PHF assembly[88]
NitrationTyr29Accumulation of oligomeric species.[89]
N-glycosylationUnder investigation. Putative sites: Asn167, Ans359, Asn410 Higher levels in AD patients’ brains. Promotion of tau hyperphosphorylation and PHF accumulation.[83,90]
O-glycosylationUnder investigationLower levels in AD patients’ brains. It prevents tau hyperphosphorylation and PHF formation and PHF accumulation.[91]
UbiquitinationLys48, Lys63It has been proposed to contribute to the formation of the tangles.[92,93,94]
α-SynFragmentationC-terminally truncated α-syn of around 10–15 kDaAccelerated aggregation.[28]
PhosphorylationSer129Enhanced aggregation and toxicity.[95,96]
AcetylationN-terminalIncreased helical propensity, altered fibril polymorphism and decreased aggregation.[97]
OxidationMet1, Met5, Met116, Met127Inhibition of fibrillation by stabilization of soluble oligomeric species.[98]
NitrationTyr39Enhanced aggregation.[99]
O-glycosylationUnder investigation.Inhibition of aggregation.[100]
UbiquitinationLys6, Lys12, Lys21, Lys43Inhibition of aggregation.[28]
Lys10, Lys23Faster fibril formation.[28]
TDP-43FragmentationC-terminally truncated TDP-43 at 25 kDa and 35 kDa fragmentsEnhanced aggregation, altered RNA processing, and cellular redistribution.[101,102]
PhosphorylationSer409, Ser410Enhanced aggregation and cellular mislocalization.[29,75]
AcetylationLys145, Lys192Impaired RNA binding and mitochondrial function; enhanced aggregation of phosphorylated TDP-43.[29]
OxidationCys3, Cys50, Cys173, Cys175Enhanced oligomerization and self-association.[29]
UbiquitinationLys 48, Lys 63Enhanced cytoplasmic accumulation to higher molecular weight aggregates.[29]

4.2. PTMs Associated with Neurodegeneration

As previously mentioned, PTMs affect the aggregation propensity of amyloidogenic proteins and play a critical role in the pathological mechanism of neurodegenerative diseases [27,28]. Furthermore, PTMs are also found in biological samples from patients [27,28,29]. Aberrant PTMs may occur well before clinically observable symptoms. This observation makes PTMs potentially suitable to be used as biomarkers for early diagnosis (Table 1).

4.2.1. Fragmentation and Cleavage

Protein cleavage is a PTM commonly associated with amyloid aggregation and has been extensively investigated both in vitro and in vivo. In AD, for example, several APP fragments have been linked to the disease, Aβ42 and Aβ40 being the most extensively investigated ones. However, other forms of Aβ also hold a diagnostic potential, including Aβ37, Aβ38, Aβ39, and Aβ43 [32].
It has been found that the CSF concentration of Aβ42 is lower in AD patients with respect to controls in approximately 50% of cases [78]. However, currently, the CSF concentration ratio Aβ42/Aβ40 is considered diagnostically more accurate, as it takes into account the physiological inter-individual protein expression fluctuations [78,103]. Besides Aβ40 and Aβ42, the concentration of Aβ43 is lower in the CSF of patients with early-onset AD, but also in patients with mild cognitive impairment and other forms of dementia [79]. On the contrary, Aβ38 is present at higher concentrations in AD patients and specifically discriminates AD from other forms of dementia [66]. Thus, the ratio Aβ42/Aβ38 may correlate with different types of dementia and be used in combined detection diagnostic approaches [103,104].
Despite α-syn and TDP-43 also being able to undergo fragmentation, which plays a role in their pathological mechanisms, very few studies had detected truncated α-syn and TDP-43 isoforms in body fluids and their roles as potential biomarkers remained unknown [28,54,105,106].

4.2.2. Phosphorylation

In Aβ, the phosphorylation of Ser8 and Ser26 is believed to increase the abundance and stability of toxic aggregates. Enrichment of these modifications in oligomeric Aβ species, compared to monomers, have been shown using phosphor-specific mono- or polyclonal antibodies [80,81].
As a consequence of hyper-phosphorylation, tau dissociates from the microtubule network and aggregates. Therefore, tau phosphorylation is considered a key event in the formation of NFT [27]. In the recent years, several antibodies have been developed to specifically recognize p-tau and a variety of ELISA protocols have been developed for different phosphorylated epitopes. Recently, p-tau at Thr217 (p-tau217) was shown to be a robust plasma [86] and CSF [107] biomarker for AD as it distinguished AD from other neurodegenerative diseases including other tauopathies.
Similarly, α-syn is also phosphorylated at multiple residues. Noteworthy to mention is the variant of α-syn carrying the phosphorylation of Ser129 (p-α-syn129). This modification occurs by multiple kinases and affects the aggregation and the toxicity of α-syn both in vitro [95,96] and in vivo [108]. Mass spectrometry (MS) and antibody-based experiments have found that LBs isolated from PD patients’ brains are enriched in p-α-syn129. They have also shown that the concentration of p-α-syn129 in body fluids is higher in PD patients and correlates with the disease severity [109,110]. The quantification of p-α-syn129, along with total α-syn, in the CSF was suggested to aid the modeling of PD and potentially the differential diagnosis of PD from other Parkinsonisms [110,111].
Several antibodies that are specific for phosphorylated TDP-43 (e.g., at Ser409 and Ser410) have been developed. However, TDP-43 undergoes several other modifications, and probing only the phosphorylation state of the protein does not accurately describe the disease state. Detection approaches currently under development involve the combined use of different antibodies targeting different modifications and conformations of TDP-43 [75,77].

4.2.3. Acetylation

Acetylation is another major PTM involved in AD. In fact, it has been reported that the overall acetylation levels of Aβ42- and tau- aggregates are significantly higher in AD hippocampus compared to healthy subjects [112]. Furthermore, several studies have shown that the acetylation of Lys16 and Lys28 of Aβ both alter the aggregation of the peptide in vitro [38]. In particular, the acetylation of Lys16 leads to the formation of amorphous aggregates instead of amyloids [38,39].
Tau undergoes acetylation at various residues. In particular, acetylation at Lys174, Lys274 and Lys280 have received the most attention as these were shown to also regulate the function of the protein [87]. Among these modifications, Lys280 was specifically found acetylated in transgenic mice and patients’ hippocampus tissues [87].
In the case of PD and dementia with LBs (DLB), it has been reported that brain tissues (temporal cortex and dorsolateral prefrontal cortex) from patients contain soluble and aggregated N-terminally acetylated α-syn [113]. N-terminal acetylation has been found to significantly alter the structure of monomeric α-syn in vitro. This PTM induces the helical folding of the N-terminus of α-syn [97], which has been suggested to promote the pathological association of the protein to membranes [114].
TDP-43 is another protein that undergoes acetylation. This PTM abrogates RNA binding and promotes the aggregation of the protein. In particular, acetylated TDP-43 at Lys145 could be detected in lesions in ALS but not FTD patients, suggesting that TDP-43 acetylation was linked to ALS pathogenesis and could be used for discriminating ALS from FTD, which is helpful as a biomarker if easily detectable in body fluids [29].

4.2.4. Oxidation and Nitration

Oxidative stress has been described as a contributing factor in neurodegenerative diseases [115]. In AD, oxidation of Aβ at Met35 was detected post-mortem in the occipital cortex in AD patients [116]. However, this modification was shown to slow down the aggregation of the protein in vitro [30,82] and further investigations will be needed to clarify the exact role in the disease. Instead of targeting oxidized amyloidogenic proteins, current diagnostic approaches mainly focus on measuring oxidative stress biomarkers (e.g., oxidation products and specific metabolites) [115]. However, these biomarkers are not specific for neurodegeneration, as oxidative stress is also induced by inflammation and in the context of various pathologies, including cancer [115]. The development of antibodies directed against oxidized amyloidogenic proteins, such as tau and TDP-43, coupled with highly sensitive approaches may help diagnosis.
Nitration of Tyr residues has been found in several amyloidogenic proteins from patients’ samples (e.g., Tyr10 in Aβ, Tyr29 in tau, Tyr39 in α-syn) [28,30,37,89]. In particular, it has been found that nitrated α-syn levels in peripheral blood cells (monocytes and erythrocytes) are specifically higher in PD patients. Thus, this modification is currently regarded as a potential diagnostic biomarker, although larger scale studies are required [99,117]. Nitrosative stress molecules are currently assessed as potential biomarkers. However, also in this case, they are not specific for neurodegeneration and further investigation is needed [118].

4.2.5. Glycosylation

Studies found short Aβ O-glycopeptides (Aβ1-15 to 20) in AD CSF samples and suggested that they could be useful biomarkers, and the glycopeptide profiles and concentrations need to be confirmed in much larger prospective studies [119] (Table 1).
Tau has also been shown to undergo N- and O-glycosylation. Interestingly, O-glycosylation plays a protective mechanism against hyperphosphorylation and PHF formation [91]. In fact, lower levels of O-glycosylated tau are found in AD brains compared to healthy controls [91]. In contrast, N-glycosylation was shown to facilitate tau hyperphosphorylation, thus promoting its accumulation in PHF. N-glycosylation has been found in human AD brains, but not in control brains, which could be a relevant modification for diagnosis [83]. In PD, O-linked glycosylated with N-acetylglucosamine of α-syn was also found in vivo [100].
Diagnostic approaches based on the detection of glycosylated amyloidogenic proteins are still largely unexplored and new techniques are under development. The assessment of glycosylation patterns in patients include N-glycome profiling using glycoblotting and MS [120,121].

4.2.6. Ubiquitination

Ubiquitin labels misfolded proteins and targets them for degradation through the ubiquitin-proteasome system. It is believed that abnormal accumulation of amyloids might result from the alteration of the ubiquitin-proteasome pathway [122].
It has been found that ubiquitin is present at higher concentrations in AD patients’ cortical tissues compared to healthy controls [122,123]. Although early studies have shown that the majority of the ubiquitination occurs when tau is aggregated in the tangles, ubiquitin has also been found in soluble tau, for example, via Lys63 conjugation [92,93]. Notably, Lys48 and, particularly, Lys63-linked polyubiquitination, as well as monoubiquitin modifications were found to contribute to the biogenesis of the tangles. Further analyses could focus on the detection of Lys63 ubiquitinated tau to evaluate its diagnostic value [94].
α-Syn is found, mono-, di- and tri-ubiquitinated, in LBs from the cortical tissues of autopsied DLB patients’ brains [124]. While ubiquitination at Lys6, Lys12, Lys21, Lys43, and Lys96 has been shown to have an inhibitory effect on fibril formation, ubiquitination at Lys10 and Lys23 has the opposite effect and could have a pathological role. However, their presence in body fluids and connection with the PD pathology remain unknown [28].
Ubiquitinated TDP-43 is a major component of the aggregates in about 50% of FTD cases and of 97% of ALS cases, and appears to be downstream of phosphorylation. Nevertheless, the effects of ubiquitination on TDP-43 aggregation still need to be further clarified [29]. Currently, potential diagnostic approaches focus on measuring free nonconjugated ubiquitin rather than ubiquitinated proteins in the CSF [125]. It has been observed that the amount of ubiquitin in the CSF reflects the amount of protein aggregates in the brain and could serve as biomarker [125,126].

4.3. Amyloid Aggregates and Amyloid-Sensitive Probes

Currently, increasing research effort is focusing on generating biomolecular tools to target specific aggregate conformations (Figure 2). The rationale underlying this strategy consists of the fact that amyloidogenic proteins are generally poorly toxic when monomeric, while their toxicity significantly increases when they are aggregated [13]. Recent evidence shows that oligomers are particularly toxic and [13], thus, in order to monitor the progression and severity of neurodegenerative diseases, one could specifically quantify the concentration of the amyloids and/or of the oligomers (Table 2, Figure 3).
In this context, several probes for amyloids and amyloid-like aggregates have been developed, including small molecule fluorescent probes, conformation-selective antibodies and aptamers [127,128,129,130].
In particular, single-molecule studies using amyloid-sensitive fluorescent molecules were able to probe individual soluble aggregates present in body fluids of patients. To do so, small molecules, such as thioflavin T (ThT), which increase their fluorescence upon binding to the cross-β-sheet component of the amyloids, have been used. Using this approach, researchers managed to quantify soluble amyloid-like aggregates in AD and PD CSF samples [131,132]. Besides ThT, other molecules with similar fluorescent properties have been developed, including molecular rotors [133,134], such as thioflavin X [135].
In addition to these small molecule probes, high-specificity biomolecules have been developed, such as antibody fragments and aptamers able to probe oligomers of different amyloidogenic proteins [127,136,137]. Recently, super-resolution microscopy with amyloid-specific aptamer and DNA point accumulation in nanoscale topography was able to detect aggregated α-syn and Aβ in samples from PD patients without the need of a conjugated fluorophore [138].
Traditionally, antibodies have been generated using well-established techniques in vivo by immunization and hybridoma technologies [139,140] or in vitro using display technologies [141]. The emergence of novel in silico antibody discovery methods have allowed the generation of antibodies in a cost- and time-effective manner [142]. AbDesign, a structure-based algorithm that utilizes combinatorial and energy-based design, has been successful in designing antibody fragments that target insulin and mycobacterial acyl-carrier protein [143]. Similar successes have been made using structure-based procedures such as OptMAVEn [144] and Rosetta antibody design [145]. More recently, the Cascade method [128] has been developed to design complementary peptides to a target epitope that can be grafted to the complementarity-determining region of an antibody scaffold. The Cascade method has previously been employed in designing conformation-specific antibodies to target Aβ42 amyloids and oligomers [127,146].

4.4. PET Neuroimaging of Amyloids in the Brain

PET imaging utilizes radiotracers to screen the presence of amyloids in the brain in vivo [147]. The radiotracers are characterized by high lipophilicity, which allows them to pass across the Blood-Brain-Barrier. Pittsburgh compound B (11C-PiB) is a fluorescent analog of ThT and was first used to trace amyloid fibrils using PET in 2004. 11C-PiB is currently among the most used radio ligands for PET imaging of cerebral Aβ pathology [148] (Table 2). Amyloid-PET can distinguish dementia from mild cognitive impairment and normal aging. However, it cannot discriminate different forms of dementia. Furthermore, these techniques are expensive and current clinically used radiotracers only reflect the level of insoluble aggregates in the brain. In contrast, novel antibody-based radioligands are under development to detect soluble oligomeric and protofibrillar Aβ forms and have been tested in transgenic mice [149,150].

4.5. Combined Detection

Neurodegenerative diseases share many symptoms and pathological mechanisms. In particular, aggregates of the same amyloidogenic protein can exist in people affected by different forms of neurodegeneration [151]. For example, Aβ and p-tau aggregates can also be found in patients with DLB, and α-syn aggregates in patients with AD. The co-occurrence of Aβ, α-syn, and tau suggests an overlap between AD, tauopathies and synucleinopathies [152]. Amyloid aggregates can also be present in people who do not show any disease symptoms [151]. These observations lead to the conclusion that, in some cases, monitoring one individual amyloidogenic protein alone may not be sufficient for delivering an accurate diagnosis.
Various studies have assessed the diagnostic performance of monitoring the levels of different amyloidogenic proteins together (Table 2). For example, the quantification of the CSF concentrations of both p-tau181, carrying the phosphorylated Thr181, and the ratio Aβ42/Aβ38 was able to differentiate AD from other neurodegenerative diseases [66,104]. A recent study conducted on a cohort of 4444 participants over a period of 14 years was able to associate the plasma levels of NFTs and Aβ42 with the risk of developing AD and all-cause dementia [153]. Furthermore, the analysis of the CSF levels of both total α-syn and total tau could help the identification of synucleinopathies over other neurodegenerative diseases [68]. A recent work has shown that the determination of the ratio of oligomeric-α-syn/total α-syn, p-α-syn129, and p-tau181 in the CSF was able to identify PD patients from controls [154]. It has also been shown that an increase in glycation, α-syn Tyr 39 nitration and pTyr125, and a decrease in SUMO-1 levels in blood samples was associate with PD [117]. Finally, it has been found that the concentration in the CSF of total TDP-43 and the ratio total tau/pThr181-tau discriminate ALS/FTD patients from healthy controls [155]. In Table 2, we summarize the main proteins which can be monitored in combination for diagnostic purposes.
Table 2. Main current and potential diagnostic markers of neurodegenerative diseases with main references.
Table 2. Main current and potential diagnostic markers of neurodegenerative diseases with main references.
Type of Diagnostic MarkersKey RemarksRefs.
Genetic
ProteinGeneMutationsPathological implications
APPAPPE693Q, A692GEarly onset familial AD[35,36]
α-SynSNCAA53T, G51D, H50Q, E46K, A30P, locus amplificationEarly onset familial PD[46,47,48,156]
TDP-43TARDBPG294A, Q331K, M337V, K181ESporadic and familial ALS[58,59]
FUSFUSR521C, R521HEarly onset ALS[63]
Neuroimaging
Protein speciesAnalytical technique
Aβ and tau aggregatesPET with 18F-FDDNP. Lacks protein specific and is unable to distinguish different forms of neurodegeneration.[147]
Tau aggregatesPET with 18F-AV1451 or 18F-GTP1. High affinity for tau aggregates. Not able to distinguish different forms of tauopathies.[147]
Aβ aggregatesPET with 11C-PiB and Florbetapir (18F). High affinity for Aβ plaques. Not able to distinguish different forms of neurodegeneration.[147,148]
Biomarkers
Protein variantsAnalytical techniques
CSF Aβ42Immunoassays. In the majority of cases, it is present at lower concentration in patients affected by AD and other forms of dementia. It is not accurate in distinguishing different forms of neurodegeneration.[78]
CSF Aβ42/Aβ40 ratioImmunoassays. Slightly better diagnostic performance for AD than Aβ42 alone. Usually, it is used in combination with other potential biomarkers (e.g., tau).[78,103]
CSF Aβ42/Aβ38 ratio Immunoassays. Better diagnostic performance for AD than Aβ42 alone. Also used in combination with other potential biomarkers (e.g., p-tau).[78,103,107]
CSF total tauImmunoassays. Generally present at higher concentrations in the CSF of AD patients. It is not disease-specific when used alone and usually measured in association with other amyloidogenic proteins (e.g., Aβ42).[67]
CSF p-tau217, p-tau181Immunoassays and MS. Their concentrations correlate with AD. P-tau181 is generally used in combination with other potential biomarkers, including total tau and Aβ42, or with the Aβ42/Aβ38 ratio.[107]
Plasma p-tau217Immunoassays and MS. Able to discriminate AD from other forms of neurodegeneration.[86]
CSF and plasma p-α-syn129Immunoassays. Good diagnostic performance for PD when used in combination with other potential biomarkers, including tau and α-syn oligomers.[109,110,154]
CSF Aβ oligomers and α-syn oligomersImmunoassays and single-molecule approaches. Good potential diagnostic performance for AD or PD, respectively. However, their use as biomarkers is not widely implemented yet. Some works have used them in combination with other potential biomarkers.[109,132,154]

5. Recent Advances in Detection Technology

Given their affordable costs and the possibility of easily making them high-throughput, immunoassays, such as ELISA and immunoblotting, have been the most commonly used techniques to quantify amyloidogenic proteins’ concentrations in body fluids and tissues. Other approaches are also in use, including PET, MS, and microscopy, and novel detection technologies with ultra-sensitivity are emerging.
An important example is provided by MS. A recently established capillary isotachophoresis–electrospray ionization MS could detect picomolar concentrations of Aβ [157]. In another study, an automated clinical mass spectrometer could detect different Aβ variants in the CSF in a multiplex manner [158]. Noteworthy, both approaches were antibody-free and did not require an immuno-enrichment step. Aβ42/Aβ40 ratios in CSF could also be determined by LC-MS/MS assay with a high clinical sensitivity [159]. The benefits of MS include small sample size, fast turnaround time, broad applicability and sensitivity.
Single-particle analysis of amyloids can be performed by microscopic methods including fluorescence, atomic force, and electron microscopy. Notably, fluorescence-based methods provide ultrasensitive detection of individual amyloid fibrils and oligomers in neurodegenerative diseases. Furthermore, super-resolution methods offer insight into structural properties and surface hydrophobicity [17].
Nanopore sensing is a non-optical technique that has recently been demonstrated to allow single-molecule analysis of polymeric proteins and could be extended to amyloids and oligomers [160,161]. In nanopore sensing, a biomolecule is translocated through a nanopore embedded within a thin dielectric membrane separating two chambers with electrolytes. Distinct conformations of the biomolecule can be characterized upon its translocation through the nanopore by the analysis of the change in the ionic conductance of the pore [162,163].
Other recent methods include amyloid seeding assays, such as protein misfolding cyclic amplification [164], and real-time quaking-induced conversion (RT-QuIC) [165]. In these assays, the composition and number of amyloids in biological samples is determined by the ability of these samples to induce the aggregation of a recombinant monomeric amyloidogenic protein using ThT-based aggregation measurements. These assays were established for several amyloidogenic proteins. Especially, RT-QuIC assay for α-syn showed high diagnostic sensitivity for PD and DLB [165].
Both MS and microscopic techniques require sophisticated instrumentation. ELISA kits for several biomarkers including Aβ42, p-tau, total-tau and α-syn are commercially available. However, ELISA can be labor-intensive to set up and with confined sensitivity. Various high sensitivity ELISA techniques have been developed. For instance, researchers have measured total α-syn concentration in body fluids using immobilized lipids [166]. Also, ELISA has been coupled with other detection technologies, such as novel plate-based electrochemiluminescence [167]. This approach achieved markedly shortened processing time, with smaller sample volume requirements and simultaneous processing of multiple biomarkers [167]. Besides electrochemiluminescence, digital ELISA has also been developed with single molecule array (Simoa) technology. This method is reported to have increased sensitivity for Aβ42 detection in the human plasma (in the pM range) [168]. Moreover, a surface-based fluorescence intensity distribution analysis (sFIDA) assay was established resembling a sandwich ELISA where Aβ oligomers were immobilized on the functionalized glass surface via antibodies, imaged by high-resolution fluorescence microscopy [169].
The Multi-Analyte Profiling (xMAP) platform stands out from a wide range of approaches based on its multiplexing capability. Simultaneous quantification of up to 100 samples in a single assay could be achieved on a semi-automated assay. Studies provided that xMAP data for total tau, p-tau, Aβ40 and Aβ42 correlated well with research-based ELISA values with higher sensitivity and specificity [170].
Immuno-polymerase chain reaction (I-PCR) utilizes real-time PCR (also known as quantitative PCR) to combine nucleic acid amplification with antibody-based assays to increase 10 to 109-fold sensitivity of conventional immune assays. Researchers quantified multiple phosphorylated tau epitopes using I-PCR in CSF [171] and developed a nano-I-PCR approach which utilized gold nanoparticles functionalized with a tau-specific monoclonal antibody for total tau quantification in CSF samples [172]. The level of total Aβ40 present in microdissected neurons could also be quantitated using I-PCR with high sensitivity and detection range [173]. This technique is suitable for small sample volumes, provides rapid time to results and can be amenable to multiplexing.
Point-of-care (POC) diagnosis is, undoubtedly, an emerging trend. This approach allows conventional ELISA, Luminex xMAP and qPCR to be developed into inexpensive, portable and easy-to-use POC devices [174]. Paper-based ELISA is the simplest option for POC diagnostics.

6. Conclusions and Potential Future Directions

Disease biomarkers represent an essential requirement for the development of accurate diagnostic approaches. In this review, we have discussed how amyloidogenic proteins hold potential as biomarkers for neurodegenerative diseases and described detection technologies to assess their concentrations in the body (Figure 3). Besides genetic mutations, many PTMs and specific conformations of amyloidogenic proteins are associated with disease and are emerging as potential biomarkers (Table 1 and Table 2). In this context, noteworthy among all aggregated conformations are the oligomers, which are highly toxic and regarded as major players in the disease onset and progression.
The use of amyloidogenic proteins as biomarkers comes with challenges. Firstly, neurodegenerative diseases share some key pathological mechanisms, including the formation of aggregates by the same amyloidogenic protein (e.g., Aβ and p-tau deposits can be found in patients with DLB) [152]. This makes it difficult to distinguish one form of neurodegeneration from another based on the detection on one specific amyloidogenic protein alone. Furthermore, amyloidogenic proteins are difficult to access within the CNS and their concentrations in accessible body fluids have noticeable fluctuations, particularly at early stages of disease.
In our opinion, promising diagnostic strategies that may overcome these issues are those based on the detection of multiple amyloidogenic proteins or protein features (Section 4.5). Additionally, amyloidogenic proteins could be monitored in conjunction with other types of biomarkers, such as metabolites. Recent investigations have shown that metabolic pathways are affected in neurodegeneration, and detection platforms have been developed for metabolic profiling [175]. For examples, nuclear magnetic resonance and MS have been successfully employed to determine metabolic changes in cellular systems [176], post-mortem brain samples [177], and CSF from patients [178]. Combined approaches could also involve neuropsychological assessment and neuroimaging.
Furthermore, amyloidogenic proteins can also be detected in regions of the body besides the CNS and body fluids. For example, aggregated forms of α-syn have been found in the digestive system of PD patients [179]. The diagnostic relevance of this finding is twofold: it shows that other parts of the body can be examined for potential biomarkers of neurodegeneration [180]; it also implies that patients may show symptoms/disorders that are apparently unrelated with the neurodegenerative condition but instead could be used for early diagnosis.
Sensitivity and specificity are important attributes of detection technologies for amyloidogenic proteins. In Section 4.3 and Section 5, we described several promising approaches which are currently under development. These are based on biosensors, single-molecule detection, and molecular probes (e.g., antibodies). In our view, antibody-based approaches in particular hold a great potential as they allow detection in complex mixtures. Moreover, antibodies can be developed to target different protein features, including PTMs and conformations (e.g., the oligomers).
In conclusion, amyloidogenic proteins are appealing potential biomarkers of neurodegeneration. Their diagnostic success is intertwined with the development of combined detection strategies, involving other types of biomarkers, organ systems, and ultra-sensitive technologies.

Author Contributions

Writing—original draft preparation, Y.J., D.M.V., D.G., Y.G., R.T., J.T.W., F.A.A.; writing—review and editing, Y.J., D.M.V., D.G., Y.G., R.T., J.T.W., F.A.A. All authors have read and agreed to the published version of the manuscript.

Funding

We thank UK Research and Innovation (Future Leaders Fellowship MR/S033947/1), the Alzheimer’s Society, UK (Grant 511), and Alzheimer’s Research UK (ARUK-PG2019B-020) for support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Figures were created with ChemDraw v19.1 and Biorender.com (accessed on 5 April 2021).

Conflicts of Interest

F.A.A. is an inventor of patent PCT/GB2020/051965.

References

  1. Graham, M.E. The securitisation of dementia: Socialities of securitisation on secure dementia care units. Ageing Soc. 2021, 41, 439–455. [Google Scholar] [CrossRef]
  2. Agrawal, M.; Biswas, A. Molecular diagnostics of neurodegenerative disorders. Front. Mol. Biosci. 2015, 2, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hussain, R.; Zubair, H.; Pursell, S.; Shahab, M. Neurodegenerative diseases: Regenerative mechanisms and novel therapeutic approaches. Brain Sci. 2018, 8, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Reul, S.; Lohmann, H.; Wiendl, H.; Duning, T.; Johnen, A. Can cognitive assessment really discriminate early stages of Alzheimer’s and behavioural variant frontotemporal dementia at initial clinical presentation? Alzheimers Res. Ther. 2017, 9, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Van Steenoven, I.; Koel-Simmelink, M.J.; Vergouw, L.J.; Tijms, B.M.; Piersma, S.R.; Pham, T.V.; Bridel, C.; Ferri, G.-L.; Cocco, C.; Noli, B.; et al. Identification of novel cerebrospinal fluid biomarker candidates for dementia with Lewy bodies: A proteomic approach. Mol. Neurodegener. 2020, 15, 36. [Google Scholar] [CrossRef] [PubMed]
  6. Li, B.; Ge, P.; Murray, K.A.; Sheth, P.; Zhang, M.; Nair, G.; Sawaya, M.R.; Shin, W.S.; Boyer, D.R.; Ye, S.; et al. Cryo-EM of full-length α-synuclein reveals fibril polymorphs with a common structural kernel. Nat. Commun. 2018, 9, 3609. [Google Scholar] [CrossRef]
  7. Dobson, C.M. Protein folding and misfolding. Nature 2003, 426, 884–890. [Google Scholar] [CrossRef]
  8. Knowles, T.P.; Vendruscolo, M.; Dobson, C.M. The amyloid state and its association with protein misfolding diseases. Nat. Rev. Mol. Cell Biol. 2014, 15, 384–396. [Google Scholar] [CrossRef] [PubMed]
  9. Astoricchio, E.; Alfano, C.; Rajendran, L.; Temussi, P.A.; Pastore, A. The Wide World of Coacervates: From the Sea to Neurodegeneration. Trends Biochem. Sci. 2020, 45, 706–717. [Google Scholar] [CrossRef]
  10. Buratti, E.; Baralle, F.E. TDP-43: Gumming up neurons through protein–protein and protein–RNA interactions. Trends Biochem. Sci. 2012, 37, 237–247. [Google Scholar] [CrossRef] [PubMed]
  11. Vogler, T.O.; Wheeler, J.R.; Nguyen, E.D.; Hughes, M.P.; Britson, K.A.; Lester, E.; Rao, B.; Dalla Betta, N.; Whitney, O.N.; Ewachiw, T.E.; et al. TDP-43 and RNA form amyloid-like myo-granules in regenerating muscle. Nature 2018, 563, 508–513. [Google Scholar] [CrossRef]
  12. Bhardwaj, A.; Myers, M.P.; Buratti, E.; Baralle, F.E. Characterizing TDP-43 interaction with its RNA targets. Nucleic Acids Res. 2013, 41, 5062–5074. [Google Scholar] [CrossRef] [Green Version]
  13. Bucciantini, M.; Rigacci, S.; Stefani, M. Amyloid aggregation: Role of biological membranes and the aggregate–membrane system. J. Phys. Chem. Lett. 2014, 5, 517–527. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, S.W.; Drakulic, S.; Deas, E.; Ouberai, M.; Aprile, F.A.; Arranz, R.; Ness, S.; Roodveldt, C.; Guilliams, T.; De-Genst, E.J.; et al. Structural characterization of toxic oligomers that are kinetically trapped during α-synuclein fibril formation. Proc. Natl. Acad. Sci. USA 2015, 112, E1994–E2003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ghosh, D.; Mehra, S.; Sahay, S.; Singh, P.K.; Maji, S.K. α-synuclein aggregation and its modulation. Int. J. Biol. Macromol. 2017, 100, 37–54. [Google Scholar] [CrossRef] [PubMed]
  16. Chen, G.-f.; Xu, T.-h.; Yan, Y.; Zhou, Y.-r.; Jiang, Y.; Melcher, K.; Xu, H.E. Amyloid beta: Structure, biology and structure-based therapeutic development. Acta Pharmacol. Sin. 2017, 38, 1205–1235. [Google Scholar] [CrossRef]
  17. De, S.; Klenerman, D. Imaging individual protein aggregates to follow aggregation and determine the role of aggregates in neurodegenerative disease. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 870–878. [Google Scholar] [CrossRef]
  18. Cremades, N.; Cohen, S.I.; Deas, E.; Abramov, A.Y.; Chen, A.Y.; Orte, A.; Sandal, M.; Clarke, R.W.; Dunne, P.; Aprile, F.A.; et al. Direct observation of the interconversion of normal and toxic forms of α-synuclein. Cell 2012, 149, 1048–1059. [Google Scholar] [CrossRef] [Green Version]
  19. Ghosh, D.; Singh, P.K.; Sahay, S.; Jha, N.N.; Jacob, R.S.; Sen, S.; Kumar, A.; Riek, R.; Maji, S.K. Structure based aggregation studies reveal the presence of helix-rich intermediate during α-Synuclein aggregation. Sci. Rep. 2015, 5, 9228. [Google Scholar] [CrossRef] [Green Version]
  20. Shea, D.; Hsu, C.-C.; Bi, T.M.; Paranjapye, N.; Childers, M.C.; Cochran, J.; Tomberlin, C.P.; Wang, L.; Paris, D.; Zonderman, J.; et al. α-Sheet secondary structure in amyloid β-peptide drives aggregation and toxicity in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2019, 116, 8895–8900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Fusco, G.; Chen, S.W.; Williamson, P.T.; Cascella, R.; Perni, M.; Jarvis, J.A.; Cecchi, C.; Vendruscolo, M.; Chiti, F.; Cremades, N.; et al. Structural basis of membrane disruption and cellular toxicity by α-synuclein oligomers. Science 2017, 358, 1440–1443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. De, S.; Wirthensohn, D.C.; Flagmeier, P.; Hughes, C.; Aprile, F.A.; Ruggeri, F.S.; Whiten, D.R.; Emin, D.; Xia, Z.; Varela, J.A.; et al. Different soluble aggregates of Aβ42 can give rise to cellular toxicity through different mechanisms. Nat. Commun. 2019, 10, 1541. [Google Scholar] [CrossRef] [Green Version]
  23. Bucciantini, M.; Calloni, G.; Chiti, F.; Formigli, L.; Nosi, D.; Dobson, C.M.; Stefani, M. Prefibrillar amyloid protein aggregates share common features of cytotoxicity. J. Biol. Chem. 2004, 279, 31374–31382. [Google Scholar] [CrossRef] [Green Version]
  24. Angelova, P.R.; Abramov, A.Y. Alpha-synuclein and beta-amyloid–different targets, same players: Calcium, free radicals and mitochondria in the mechanism of neurodegeneration. Biochem. Biophys. Res. Commun. 2017, 483, 1110–1115. [Google Scholar] [CrossRef]
  25. Koffie, R.M.; Meyer-Luehmann, M.; Hashimoto, T.; Adams, K.W.; Mielke, M.L.; Garcia-Alloza, M.; Micheva, K.D.; Smith, S.J.; Kim, M.L.; Lee, V.M.; et al. Oligomeric amyloid β associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc. Natl. Acad. Sci. USA 2009, 106, 4012–4017. [Google Scholar] [CrossRef] [Green Version]
  26. Winner, B.; Jappelli, R.; Maji, S.K.; Desplats, P.A.; Boyer, L.; Aigner, S.; Hetzer, C.; Loher, T.; Vilar, M.; Campioni, S.; et al. In vivo demonstration that α-synuclein oligomers are toxic. Proc. Natl. Acad. Sci. USA 2011, 108, 4194–4199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Gong, C.-X.; Liu, F.; Grundke-Iqbal, I.; Iqbal, K. Post-translational modifications of tau protein in Alzheimer’s disease. J. Neural Transm. 2005, 112, 813–838. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, J.; Li, X.; Li, J.-D. The Roles of Post-translational Modifications on α-Synuclein in the Pathogenesis of Parkinson’s Diseases. Front. Neurosci. 2019, 13, 381. [Google Scholar] [CrossRef] [Green Version]
  29. Prasad, A.; Bharathi, V.; Sivalingam, V.; Girdhar, A.; Patel, B.K. Molecular mechanisms of TDP-43 misfolding and pathology in amyotrophic lateral sclerosis. Front. Mol. Neurosci. 2019, 12, 25. [Google Scholar] [CrossRef] [PubMed]
  30. Kummer, M.P.; Heneka, M.T. Truncated and modified amyloid-beta species. Alzheimers Res. Ther. 2014, 6, 28. [Google Scholar] [CrossRef] [Green Version]
  31. Buratti, E. TDP-43 post-translational modifications in health and disease. Expert Opin. Ther. Targets 2018, 22, 279–293. [Google Scholar] [CrossRef] [PubMed]
  32. Mayeux, R. Biomarkers: Potential uses and limitations. NeuroRx 2004, 1, 182–188. [Google Scholar] [CrossRef]
  33. Blennow, K. CSF biomarkers for Alzheimer’s disease: Use in early diagnosis and evaluation of drug treatment. Expert Rev. Mol. Diagn. 2005, 5, 661–672. [Google Scholar] [CrossRef]
  34. Hatami, A.; Monjazeb, S.; Milton, S.; Glabe, C.G. Familial Alzheimer’s disease mutations within the amyloid precursor protein alter the aggregation and conformation of the amyloid-β peptide. J. Biol. Chem. 2017, 292, 3172–3185. [Google Scholar] [CrossRef] [Green Version]
  35. Hendriks, L.; van Duijn, C.M.; Cras, P.; Cruts, M.; Van Hul, W.; van Harskamp, F.; Warren, A.; McInnis, M.G.; Antonarakis, S.E.; Martin, J.-J.; et al. Presenile dementia and cerebral haemorrhage linked to a mutation at codon 692 of the β–amyloid precursor protein gene. Nat. Genet. 1992, 1, 218–221. [Google Scholar] [CrossRef]
  36. Nilsberth, C.; Westlind-Danielsson, A.; Eckman, C.B.; Condron, M.M.; Axelman, K.; Forsell, C.; Stenh, C.; Luthman, J.; Teplow, D.B.; Younkin, S.G.; et al. The ‘Arctic’APP mutation (E693G) causes Alzheimer’s disease by enhanced Aβ protofibril formation. Nat. Neurosci. 2001, 4, 887–893. [Google Scholar] [CrossRef] [PubMed]
  37. Kummer, M.P.; Hermes, M.; Delekarte, A.; Hammerschmidt, T.; Kumar, S.; Terwel, D.; Walter, J.; Pape, H.-C.; König, S.; Roeber, S.; et al. Nitration of tyrosine 10 critically enhances amyloid β aggregation and plaque formation. Neuron 2011, 71, 833–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Pilkington IV, A.W.; Schupp, J.; Nyman, M.; Valentine, S.J.; Smith, D.M.; Legleiter, J. Acetylation of Aβ40 Alters Aggregation in the Presence and Absence of Lipid Membranes. ACS Chem. Neurosci. 2019, 11, 146–161. [Google Scholar] [CrossRef] [PubMed]
  39. Adhikari, R.; Yang, M.; Saikia, N.; Dutta, C.; Alharbi, W.F.; Shan, Z.; Pandey, R.; Tiwari, A. Acetylation of Aβ42 at Lysine 16 Disrupts Amyloid Formation. ACS Chem. Neurosci. 2020, 11, 1178–1191. [Google Scholar] [CrossRef]
  40. Oakley, S.S.; Maina, M.B.; Marshall, K.E.; Al-Hilaly, Y.K.; Harrington, C.R.; Wischik, C.M.; Serpell, L.C. Tau Filament Self-Assembly and Structure: Tau as a Therapeutic Target. Front. Neurol. 2020, 11, 1207. [Google Scholar] [CrossRef] [PubMed]
  41. Goedert, M. Tau filaments in neurodegenerative diseases. FEBS Lett. 2018, 592, 2383–2391. [Google Scholar] [CrossRef] [PubMed]
  42. Fitzpatrick, A.W.; Falcon, B.; He, S.; Murzin, A.G.; Murshudov, G.; Garringer, H.J.; Crowther, R.A.; Ghetti, B.; Goedert, M.; Scheres, S.H. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature 2017, 547, 185–190. [Google Scholar] [CrossRef] [Green Version]
  43. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.-Y.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. α-Synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef] [PubMed]
  44. Rachakonda, V.; Pan, T.H.; Le, W.D. Biomarkers of neurodegenerative disorders: How good are they? Cell Res. 2004, 14, 349–358. [Google Scholar] [CrossRef] [PubMed]
  45. Auluck, P.K.; Caraveo, G.; Lindquist, S. α-Synuclein: Membrane interactions and toxicity in Parkinson’s disease. Annu. Rev. Cell Dev. Biol. 2010, 26, 211–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Goedert, M. Alpha-synuclein and neurodegenerative diseases. Nat. Rev. Neurosci. 2001, 2, 492–501. [Google Scholar] [CrossRef]
  47. Lesage, S.; Anheim, M.; Letournel, F.; Bousset, L.; Honoré, A.; Rozas, N.; Pieri, L.; Madiona, K.; Dürr, A.; Melki, R.; et al. G51D α-synuclein mutation causes a novel Parkinsonian–pyramidal syndrome. Ann. Neurol. 2013, 73, 459–471. [Google Scholar] [CrossRef]
  48. Book, A.; Guella, I.; Candido, T.; Brice, A.; Hattori, N.; Jeon, B.; Farrer, M.J.; SNCA Multiplication Investigators of the GEoPD Consortium. A meta-analysis of α-synuclein multiplication in familial parkinsonism. Front. Neurol. 2018, 9, 1021. [Google Scholar] [CrossRef]
  49. Surgucheva, I.; Sharov, V.S.; Surguchov, A. γ-Synuclein: Seeding of α-synuclein aggregation and transmission between cells. Biochemistry 2012, 51, 4743–4754. [Google Scholar] [CrossRef]
  50. Brown, J.W.; Buell, A.K.; Michaels, T.C.; Meisl, G.; Carozza, J.; Flagmeier, P.; Vendruscolo, M.; Knowles, T.P.; Dobson, C.M.; Galvagnion, C. β-Synuclein suppresses both the initiation and amplification steps of α-synuclein aggregation via competitive binding to surfaces. Sci. Rep. 2016, 6, 36010. [Google Scholar] [CrossRef]
  51. Schmid, A.W.; Fauvet, B.; Moniatte, M.; Lashuel, H.A. Alpha-synuclein post-translational modifications as potential biomarkers for Parkinson disease and other synucleinopathies. Mol. Cell. Proteomics 2013, 12, 3543–3558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Vlad, C.; Lindner, K.; Karreman, C.; Schildknecht, S.; Leist, M.; Tomczyk, N.; Rontree, J.; Langridge, J.; Danzer, K.; Ciossek, T. Autoproteolytic fragments are intermediates in the oligomerization-aggregation of Parkinson’s disease protein alpha-synuclein as revealed by ion mobility mass spectrometry. ChemBioChem 2011, 12, 2740–2744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kellie, J.F.; Higgs, R.E.; Ryder, J.W.; Major, A.; Beach, T.G.; Adler, C.H.; Merchant, K.; Knierman, M.D. Quantitative measurement of intact alpha-synuclein proteoforms from post-mortem control and Parkinson’s disease brain tissue by intact protein mass spectrometry. Sci. Rep. 2014, 4, 5797. [Google Scholar] [CrossRef] [PubMed]
  54. Feneberg, E.; Gray, E.; Ansorge, O.; Talbot, K.; Turner, M.R. Towards a TDP-43-based biomarker for ALS and FTLD. Mol. Neurobiol. 2018, 55, 7789–7801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Kumar, V.; Prakash, A.; Tomar, A.K.; Srivastava, A.; Kundu, B.; Lynn, A.M.; Hassan, M.I. Exploring the aggregation-prone regions from structural domains of human TDP-43. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 286–296. [Google Scholar] [CrossRef]
  56. Zacco, E.; Martin, S.R.; Thorogate, R.; Pastore, A. The RNA-recognition motifs of TAR DNA-binding protein 43 may play a role in the aberrant self-assembly of the protein. Front. Mol. Neurosci. 2018, 11, 372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Zacco, E.; Graña-Montes, R.; Martin, S.R.; de Groot, N.S.; Alfano, C.; Tartaglia, G.G.; Pastore, A. RNA as a key factor in driving or preventing self-assembly of the TAR DNA-binding protein 43. J. Mol. Biol. 2019, 431, 1671–1688. [Google Scholar] [CrossRef] [PubMed]
  58. Sreedharan, J.; Blair, I.P.; Tripathi, V.B.; Hu, X.; Vance, C.; Rogelj, B.; Ackerley, S.; Durnall, J.C.; Williams, K.L.; Buratti, E.; et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 2008, 319, 1668–1672. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, H.-J.; Topp, S.D.; Hui, H.S.; Zacco, E.; Katarya, M.; McLoughlin, C.; King, A.; Smith, B.N.; Troakes, C.; Pastore, A.; et al. RRM adjacent TARDBP mutations disrupt RNA binding and enhance TDP-43 proteinopathy. Brain 2019, 142, 3753–3770. [Google Scholar] [CrossRef]
  60. McAlary, L.; Plotkin, S.S.; Yerbury, J.J.; Cashman, N.R. Prion-like propagation of protein misfolding and aggregation in amyotrophic lateral sclerosis. Front. Mol. Neurosci. 2019, 12, 262. [Google Scholar] [CrossRef] [Green Version]
  61. Nolan, M.; Talbot, K.; Ansorge, O. Pathogenesis of FUS-associated ALS and FTD: Insights from rodent models. Acta Neuropathol. Commun. 2016, 4, 99. [Google Scholar] [CrossRef] [Green Version]
  62. St George-Hyslop, P.; Lin, J.Q.; Miyashita, A.; Phillips, E.C.; Qamar, S.; Randle, S.J.; Wang, G. The physiological and pathological biophysics of phase separation and gelation of RNA binding proteins in amyotrophic lateral sclerosis and fronto-temporal lobar degeneration. Brain Res. 2018, 1693, 11–23. [Google Scholar] [CrossRef]
  63. Vance, C.; Rogelj, B.; Hortobágyi, T.; De Vos, K.J.; Nishimura, A.L.; Sreedharan, J.; Hu, X.; Smith, B.; Ruddy, D.; Wright, P.; et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 2009, 323, 1208–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Rhoads, S.N.; Monahan, Z.T.; Yee, D.S.; Shewmaker, F.P. The role of post-translational modifications on prion-like aggregation and liquid-phase separation of FUS. Int. J. Mol. Sci. 2018, 19, 886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Meeter, L.H.; Kaat, L.D.; Rohrer, J.D.; Van Swieten, J.C. Imaging and fluid biomarkers in frontotemporal dementia. Nat. Rev. Neurol. 2017, 13, 406–419. [Google Scholar] [CrossRef]
  66. Lee, J.C.; Kim, S.J.; Hong, S.; Kim, Y. Diagnosis of Alzheimer’s disease utilizing amyloid and tau as fluid biomarkers. Exp. Mol. Med. 2019, 51, 1–10. [Google Scholar] [CrossRef] [Green Version]
  67. Sunderland, T.; Linker, G.; Mirza, N.; Putnam, K.T.; Friedman, D.L.; Kimmel, L.H.; Bergeson, J.; Manetti, G.J.; Zimmermann, M.; Tang, B.; et al. Decreased β-amyloid1-42 and increased tau levels in cerebrospinal fluid of patients with Alzheimer disease. JAMA 2003, 289, 2094–2103. [Google Scholar] [CrossRef] [Green Version]
  68. Mollenhauer, B.; Locascio, J.J.; Schulz-Schaeffer, W.; Sixel-Döring, F.; Trenkwalder, C.; Schlossmacher, M.G. α-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: A cohort study. The Lancet Neurol. 2011, 10, 230–240. [Google Scholar] [CrossRef]
  69. Atik, A.; Stewart, T.; Zhang, J. Alpha-synuclein as a biomarker for Parkinson’s disease. Brain Pathol. 2016, 26, 410–418. [Google Scholar] [CrossRef]
  70. Mollenhauer, B. Quantification of α-synuclein in cerebrospinal fluid: How ideal is this biomarker for Parkinson’s disease? Parkinsonism Relat. Disord. 2014, 20, S76–S79. [Google Scholar] [CrossRef]
  71. Foulds, P.; McAuley, E.; Gibbons, L.; Davidson, Y.; Pickering-Brown, S.M.; Neary, D.; Snowden, J.S.; Allsop, D.; Mann, D.M. TDP-43 protein in plasma may index TDP-43 brain pathology in Alzheimer’s disease and frontotemporal lobar degeneration. Acta Neuropathol. 2008, 116, 141–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Verstraete, E.; Kuiperij, H.B.; van Blitterswijk, M.M.; Veldink, J.H.; Schelhaas, H.J.; Van Den Berg, L.H.; Verbeek, M.M. TDP-43 plasma levels are higher in amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. 2012, 13, 446–451. [Google Scholar] [CrossRef] [PubMed]
  73. Noto, Y.-I.; Shibuya, K.; Sato, Y.; Kanai, K.; Misawa, S.; Sawai, S.; Mori, M.; Uchiyama, T.; Isose, S.; Nasu, S.; et al. Elevated CSF TDP-43 levels in amyotrophic lateral sclerosis: Specificity, sensitivity, and a possible prognostic value. Amyotroph. Lateral Scler. 2011, 12, 140–143. [Google Scholar] [CrossRef]
  74. Majumder, V.; Gregory, J.M.; Barria, M.A.; Green, A.; Pal, S. TDP-43 as a potential biomarker for amyotrophic lateral sclerosis: A systematic review and meta-analysis. BMC Neurol. 2018, 18, 90. [Google Scholar] [CrossRef] [PubMed]
  75. Steinacker, P.; Barschke, P.; Otto, M. Biomarkers for diseases with TDP-43 pathology. Mol. Cell. Neurosci. 2019, 97, 43–59. [Google Scholar] [CrossRef]
  76. Feneberg, E.; Steinacker, P.; Lehnert, S.; Schneider, A.; Walther, P.; Thal, D.R.; Linsenmeier, M.; Ludolph, A.C.; Otto, M. Limited role of free TDP-43 as a diagnostic tool in neurodegenerative diseases. Amyotroph. Lateral Scler. Front. Degener. 2014, 15, 351–356. [Google Scholar] [CrossRef] [PubMed]
  77. Goossens, J.; Vanmechelen, E.; Trojanowski, J.Q.; Lee, V.M.; Van Broeckhoven, C.; van der Zee, J.; Engelborghs, S. TDP-43 as a possible biomarker for frontotemporal lobar degeneration: A systematic review of existing antibodies. Acta Neuropathol. Commun. 2015, 3, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Struyfs, H.; Van Broeck, B.; Timmers, M.; Fransen, E.; Sleegers, K.; Van Broeckhoven, C.; De Deyn, P.P.; Streffer, J.R.; Mercken, M.; Engelborghs, S. Diagnostic accuracy of cerebrospinal fluid amyloid-β isoforms for early and differential dementia diagnosis. J. Alzheimers Dis. 2015, 45, 813–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Lauridsen, C.; Sando, S.B.; Møller, I.; Berge, G.; Pomary, P.K.; Grøntvedt, G.R.; Salvesen, Ø.; Bråthen, G.; White, L.R. 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. 2017, 9, 210. [Google Scholar] [CrossRef] [PubMed]
  80. Kumar, S.; Rezaei-Ghaleh, N.; Terwel, D.; Thal, D.R.; Richard, M.; Hoch, M.; Mc Donald, J.M.; Wüllner, U.; Glebov, K.; Heneka, M.T.; et al. Extracellular phosphorylation of the amyloid β-peptide promotes formation of toxic aggregates during the pathogenesis of Alzheimer’s disease. EMBO J. 2011, 30, 2255–2265. [Google Scholar] [CrossRef] [Green Version]
  81. Kumar, S.; Wirths, O.; Stüber, K.; Wunderlich, P.; Koch, P.; Theil, S.; Rezaei-Ghaleh, N.; Zweckstetter, M.; Bayer, T.A.; Brüstle, O.; et al. Phosphorylation of the amyloid β-peptide at Ser26 stabilizes oligomeric assembly and increases neurotoxicity. Acta Neuropathol. 2016, 131, 525–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Friedemann, M.; Helk, E.; Tiiman, A.; Zovo, K.; Palumaa, P.; Tõugu, V. Effect of methionine-35 oxidation on the aggregation of amyloid-β peptide. Biochem. Biophys. Rep. 2015, 3, 94–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Schedin-Weiss, S.; Winblad, B.; Tjernberg, L.O. The role of protein glycosylation in Alzheimer disease. FEBS J. 2014, 281, 46–62. [Google Scholar] [CrossRef]
  84. Harigaya, Y.; Saido, T.C.; Eckman, C.B.; Prada, C.-M.; Shoji, M.; Younkin, S.G. 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. 2000, 276, 422–427. [Google Scholar] [CrossRef]
  85. Perez-Garmendia, R.; Gevorkian, G. Pyroglutamate-modified amyloid beta peptides: Emerging targets for Alzheimer s disease immunotherapy. Curr. Neuropharmacol. 2013, 11, 491–498. [Google Scholar] [CrossRef] [Green Version]
  86. Palmqvist, S.; Janelidze, S.; Quiroz, Y.T.; Zetterberg, H.; Lopera, F.; Stomrud, E.; Su, Y.; Chen, Y.; Serrano, G.E.; Leuzy, A.; et al. Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 2020, 324, 772–781. [Google Scholar] [CrossRef]
  87. Cohen, T.J.; Guo, J.L.; Hurtado, D.E.; Kwong, L.K.; Mills, I.P.; Trojanowski, J.Q.; Lee, V.M. The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat. Commun. 2011, 2, 252. [Google Scholar] [CrossRef] [Green Version]
  88. Landino, L.M.; Skreslet, T.E.; Alston, J.A. Cysteine oxidation of tau and microtubule-associated protein-2 by peroxynitrite modulation of microtubule assembly kinetics by the thioredoxin reductase system. J. Biol. Chem. 2004, 279, 35101–35105. [Google Scholar] [CrossRef] [Green Version]
  89. Reynolds, M.R.; Reyes, J.F.; Fu, Y.; Bigio, E.H.; Guillozet-Bongaarts, A.L.; Berry, R.W.; Binder, L.I. Tau nitration occurs at tyrosine 29 in the fibrillar lesions of Alzheimer’s disease and other tauopathies. J. Neurosci. 2006, 26, 10636–10645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Losev, Y.; Frenkel-Pinter, M.; Abu-Hussien, M.; Viswanathan, G.K.; Elyashiv-Revivo, D.; Geries, R.; Khalaila, I.; Gazit, E.; Segal, D. Differential effects of putative N-glycosylation sites in human Tau on Alzheimer’s disease-related neurodegeneration. Cell. Mol. Life Sci. 2021, 78, 2231–2245. [Google Scholar] [CrossRef] [PubMed]
  91. Liu, F.; Shi, J.; Tanimukai, H.; Gu, J.; Gu, J.; Grundke-Iqbal, I.; Iqbal, K.; Gong, C.-X. Reduced O-GlcNAcylation links lower brain glucose metabolism and tau pathology in Alzheimer’s disease. Brain 2009, 132, 1820–1832. [Google Scholar] [CrossRef] [Green Version]
  92. Bancher, C.; Grundke-Iqbal, I.; Iqbal, K.; Fried, V.; Smith, H.; Wisniewski, H. Abnormal phosphorylation of tau precedes ubiquitination in neurofibrillary pathology of Alzheimer disease. Brain Res. 1991, 539, 11–18. [Google Scholar] [CrossRef]
  93. Kontaxi, C.; Piccardo, P.; Gill, A.C. Lysine-directed post-translational modifications of tau protein in Alzheimer’s disease and related Tauopathies. Front. Mol. Biosci. 2017, 4, 56. [Google Scholar] [CrossRef] [PubMed]
  94. Tan, J.M.; Wong, E.S.; Kirkpatrick, D.S.; Pletnikova, O.; Ko, H.S.; Tay, S.-P.; Ho, M.W.; Troncoso, J.; Gygi, S.P.; Lee, M.K.; et al. Lysine 63-linked ubiquitination promotes the formation and autophagic clearance of protein inclusions associated with neurodegenerative diseases. Hum. Mol. Genet. 2008, 17, 431–439. [Google Scholar] [CrossRef] [PubMed]
  95. Fujiwara, H.; Hasegawa, M.; Dohmae, N.; Kawashima, A.; Masliah, E.; Goldberg, M.S.; Shen, J.; Takio, K.; Iwatsubo, T. α-Synuclein is phosphorylated in synucleinopathy lesions. Nat. Cell Biol. 2002, 4, 160–164. [Google Scholar] [CrossRef] [PubMed]
  96. Smith, W.W.; Margolis, R.L.; Li, X.; Troncoso, J.C.; Lee, M.K.; Dawson, V.L.; Dawson, T.M.; Iwatsubo, T.; Ross, C.A. α-Synuclein phosphorylation enhances eosinophilic cytoplasmic inclusion formation in SH-SY5Y cells. J. Neurosci. 2005, 25, 5544–5552. [Google Scholar] [CrossRef] [PubMed]
  97. Kang, L.; Moriarty, G.M.; Woods, L.A.; Ashcroft, A.E.; Radford, S.E.; Baum, J. N-terminal acetylation of α-synuclein induces increased transient helical propensity and decreased aggregation rates in the intrinsically disordered monomer. Protein Sci. 2012, 21, 911–917. [Google Scholar] [CrossRef] [Green Version]
  98. Glaser, C.B.; Yamin, G.; Uversky, V.N.; Fink, A.L. Methionine oxidation, α-synuclein and Parkinson’s disease. Biochim Biophys Acta Proteins Proteom 2005, 1703, 157–169. [Google Scholar] [CrossRef]
  99. Prigione, A.; Piazza, F.; Brighina, L.; Begni, B.; Galbussera, A.; DiFrancesco, J.C.; Andreoni, S.; Piolti, R.; Ferrarese, C. Alpha-synuclein nitration and autophagy response are induced in peripheral blood cells from patients with Parkinson disease. Neurosci. Lett. 2010, 477, 6–10. [Google Scholar] [CrossRef]
  100. Levine, P.M.; Galesic, A.; Balana, A.T.; Mahul-Mellier, A.-L.; Navarro, M.X.; De Leon, C.A.; Lashuel, H.A.; Pratt, M.R. α-Synuclein O-GlcNAcylation alters aggregation and toxicity, revealing certain residues as potential inhibitors of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2019, 116, 1511–1519. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, Y.-J.; Xu, Y.-F.; Cook, C.; Gendron, T.F.; Roettges, P.; Link, C.D.; Lin, W.-L.; Tong, J.; Castanedes-Casey, M.; Ash, P.; et al. Aberrant cleavage of TDP-43 enhances aggregation and cellular toxicity. Proc. Natl. Acad. Sci. USA 2009, 106, 7607–7612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Che, M.X.; Jiang, Y.J.; Xie, Y.Y.; Jiang, L.L.; Hu, H.Y. Aggregation of the 35-kDa fragment of TDP-43 causes formation of cytoplasmic inclusions and alteration of RNA processing. FASEB J. 2011, 25, 2344–2353. [Google Scholar] [CrossRef]
  103. Koychev, I.; Galna, B.; Zetterberg, H.; Lawson, J.; Zamboni, G.; Ridha, B.H.; Rowe, J.B.; Thomas, A.; Howard, R.; Malhotra, P.; et al. Aβ 42/Aβ 40 and Aβ 42/Aβ 38 Ratios Are Associated with Measures of Gait Variability and Activities of Daily Living in Mild Alzheimer’s Disease: A Pilot Study. J. Alzheimers Dis. 2018, 65, 1377–1383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Welge, V.; Fiege, O.; Lewczuk, P.; Mollenhauer, B.; Esselmann, H.; Klafki, H.-W.; Wolf, S.; Trenkwalder, C.; Otto, M.; Kornhuber, J.; et al. Combined CSF tau, p-tau181 and amyloid-β 38/40/42 for diagnosing Alzheimer’s disease. J. Neural Transm. 2009, 116, 203–212. [Google Scholar] [CrossRef]
  105. Tsuji, H.; Arai, T.; Kametani, F.; Nonaka, T.; Yamashita, M.; Suzukake, M.; Hosokawa, M.; Yoshida, M.; Hatsuta, H.; Takao, M.; et al. Molecular analysis and biochemical classification of TDP-43 proteinopathy. Brain 2012, 135, 3380–3391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Steinacker, P.; Hendrich, C.; Sperfeld, A.D.; Jesse, S.; von Arnim, C.A.; Lehnert, S.; Pabst, A.; Uttner, I.; Tumani, H.; Lee, V.M.-Y.; et al. TDP-43 in cerebrospinal fluid of patients with frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Arch. Neurol. 2008, 65, 1481–1487. [Google Scholar] [CrossRef] [Green Version]
  107. Janelidze, S.; Stomrud, E.; Smith, R.; Palmqvist, S.; Mattsson, N.; Airey, D.C.; Proctor, N.K.; Chai, X.; Shcherbinin, S.; Sims, J.R.; et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nat. Commun. 2020, 11, 1683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Feany, M.B.; Bender, W.W. A Drosophila model of Parkinson’s disease. Nature 2000, 404, 394–398. [Google Scholar] [CrossRef]
  109. Foulds, P.G.; Mitchell, J.D.; Parker, A.; Turner, R.; Green, G.; Diggle, P.; Hasegawa, M.; Taylor, M.; Mann, D.; Allsop, D. Phosphorylated α-synuclein can be detected in blood plasma and is potentially a useful biomarker for Parkinson’s disease. FASEB J. 2011, 25, 4127–4137. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, Y.; Shi, M.; Chung, K.A.; Zabetian, C.P.; Leverenz, J.B.; Berg, D.; Srulijes, K.; Trojanowski, J.Q.; Lee, V.M.-Y.; Siderowf, A.D.; et al. Phosphorylated α-synuclein in Parkinson’s disease. Sci. Transl. Med. 2012, 4, ra120–ra121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Stewart, T.; Sossi, V.; Aasly, J.O.; Wszolek, Z.K.; Uitti, R.J.; Hasegawa, K.; Yokoyama, T.; Zabetian, C.P.; Leverenz, J.B.; Stoessl, A.J.; et al. Phosphorylated α-synuclein in Parkinson’s disease: Correlation depends on disease severity. Acta Neuropathol. Commun. 2015, 3, 7. [Google Scholar] [CrossRef] [Green Version]
  112. Ayyadevara, S.; Balasubramaniam, M.; Parcon, P.A.; Barger, S.W.; Griffin, W.S.T.; Alla, R.; Tackett, A.J.; Mackintosh, S.G.; Petricoin, E.; Zhou, W.; et al. Proteins that mediate protein aggregation and cytotoxicity distinguish Alzheimer’s hippocampus from normal controls. Aging Cell 2016, 15, 924–939. [Google Scholar] [CrossRef] [PubMed]
  113. Öhrfelt, A.; Zetterberg, H.; Andersson, K.; Persson, R.; Secic, D.; Brinkmalm, G.; Wallin, A.; Mulugeta, E.; Francis, P.T.; Vanmechelen, E.; et al. Identification of novel α-synuclein isoforms in human brain tissue by using an online nanoLC-ESI-FTICR-MS method. Neurochem. Res. 2011, 36, 2029–2042. [Google Scholar] [CrossRef] [Green Version]
  114. Vinueza-Gavilanes, R.; Íñigo-Marco, I.; Larrea, L.; Lasa, M.; Carte, B.; Santamaría, E.; Fernández-Irigoyen, J.; Bugallo, R.; Aragón, T.; Aldabe, R.; et al. N-terminal acetylation mutants affect alpha-synuclein stability, protein levels and neuronal toxicity. Neurobiol. Dis. 2020, 137, 104781. [Google Scholar] [CrossRef]
  115. Collin, F.; Cheignon, C.; Hureau, C. Oxidative stress as a biomarker for Alzheimer’s disease. Biomark. Med. 2018, 12, 201–203. [Google Scholar] [CrossRef] [PubMed]
  116. Näslund, J.; Schierhorn, A.; Hellman, U.; Lannfelt, L.; Roses, A.D.; Tjernberg, L.O.; Silberring, J.; Gandy, S.E.; Winblad, B.; Greengard, P. Relative abundance of Alzheimer A beta amyloid peptide variants in Alzheimer disease and normal aging. Proc. Natl. Acad. Sci. USA 1994, 91, 8378–8382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Miranda, H.V.; Cássio, R.; Correia-Guedes, L.; Gomes, M.A.; Chegão, A.; Miranda, E.; Soares, T.; Coelho, M.; Rosa, M.M.; Ferreira, J.J.; et al. Posttranslational modifications of blood-derived alpha-synuclein as biochemical markers for Parkinson’s disease. Sci. Rep. 2017, 7, 13713. [Google Scholar] [CrossRef] [Green Version]
  118. Mangialasche, F.; Polidori, M.C.; Monastero, R.; Ercolani, S.; Camarda, C.; Cecchetti, R.; Mecocci, P. Biomarkers of oxidative and nitrosative damage in Alzheimer’s disease and mild cognitive impairment. Ageing Res. Rev. 2009, 8, 285–305. [Google Scholar] [CrossRef] [Green Version]
  119. Halim, A.; Brinkmalm, G.; Rüetschi, U.; Westman-Brinkmalm, A.; Portelius, E.; Zetterberg, H.; Blennow, K.; Larson, G.; Nilsson, J. Site-specific characterization of threonine, serine, and tyrosine glycosylations of amyloid precursor protein/amyloid β-peptides in human cerebrospinal fluid. Proc. Natl. Acad. Sci. USA 2011, 108, 11848–11853. [Google Scholar] [CrossRef] [Green Version]
  120. Gizaw, S.T.; Ohashi, T.; Tanaka, M.; Hinou, H.; Nishimura, S.-I. Glycoblotting method allows for rapid and efficient glycome profiling of human Alzheimer’s disease brain, serum and cerebrospinal fluid towards potential biomarker discovery. Biochim. Biophys. Acta Gen. Subj. 2016, 1860, 1716–1727. [Google Scholar] [CrossRef] [Green Version]
  121. Palmigiano, A.; Barone, R.; Sturiale, L.; Sanfilippo, C.; Bua, R.O.; Romeo, D.A.; Messina, A.; Capuana, M.L.; Maci, T.; Le Pira, F.; et al. CSF N-glycoproteomics for early diagnosis in Alzheimer’s disease. J. Proteom. 2016, 131, 29–37. [Google Scholar] [CrossRef]
  122. Cole, G.M.; Timiras, P.S. Ubiquitin-protein conjugates in Alzheimer’s lesions. Neurosci. Lett. 1987, 79, 207–212. [Google Scholar] [CrossRef]
  123. Riederer, I.M.; Schiffrin, M.; Kövari, E.; Bouras, C.; Riederer, B.M. Ubiquitination and cysteine nitrosylation during aging and Alzheimer’s disease. Brain Res. Bull. 2009, 80, 233–241. [Google Scholar] [CrossRef]
  124. Hasegawa, M.; Fujiwara, H.; Nonaka, T.; Wakabayashi, K.; Takahashi, H.; Lee, V.M.-Y.; Trojanowski, J.Q.; Mann, D.; Iwatsubo, T. Phosphorylated α-synuclein is ubiquitinated in α-synucleinopathy lesions. J. Biol. Chem. 2002, 277, 49071–49076. [Google Scholar] [CrossRef] [Green Version]
  125. Sjödin, S.; Hansson, O.; Öhrfelt, A.; Brinkmalm, G.; Zetterberg, H.; Brinkmalm, A.; Blennow, K. Mass spectrometric analysis of cerebrospinal fluid ubiquitin in Alzheimer’s disease and Parkinsonian disorders. Proteom. Clin. Appl. 2017, 11, 1700100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Iqbal, K.; Grundke-Iqbal, I. Elevated levels of τ and ubiquitin in brain and cerebrospinal fluid in Alzheimer’s disease. Int. Psychogeriatr. 1997, 9, 289–296. [Google Scholar] [CrossRef] [PubMed]
  127. Aprile, F.A.; Sormanni, P.; Podpolny, M.; Chhangur, S.; Needham, L.-M.; Ruggeri, F.S.; Perni, M.; Limbocker, R.; Heller, G.T.; Sneideris, T.; et al. Rational design of a conformation-specific antibody for the quantification of Aβ oligomers. Proc. Natl. Acad. Sci. USA 2020, 117, 13509–13518. [Google Scholar] [CrossRef] [PubMed]
  128. Sormanni, P.; Aprile, F.A.; Vendruscolo, M. Rational design of antibodies targeting specific epitopes within intrinsically disordered proteins. Proc. Natl. Acad. Sci. USA 2015, 112, 9902–9907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Meli, G.; Lecci, A.; Manca, A.; Krako, N.; Albertini, V.; Benussi, L.; Ghidoni, R.; Cattaneo, A. Conformational targeting of intracellular Aβ oligomers demonstrates their pathological oligomerization inside the endoplasmic reticulum. Nat. Commun. 2014, 5, 3867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Perchiacca, J.M.; Ladiwala, A.R.A.; Bhattacharya, M.; Tessier, P.M. Aggregation-resistant domain antibodies engineered with charged mutations near the edges of the complementarity-determining regions. Protein Eng. Des. Sel. 2012, 25, 591–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Horrocks, M.H.; Lee, S.F.; Gandhi, S.; Magdalinou, N.K.; Chen, S.W.; Devine, M.J.; Tosatto, L.; Kjaergaard, M.; Beckwith, J.S.; Zetterberg, H.; et al. Single-molecule imaging of individual amyloid protein aggregates in human biofluids. ACS Chem. Neurosci. 2016, 7, 399–406. [Google Scholar] [CrossRef] [Green Version]
  132. De, S.; Whiten, D.R.; Ruggeri, F.S.; Hughes, C.; Rodrigues, M.; Sideris, D.I.; Taylor, C.G.; Aprile, F.A.; Muyldermans, S.; Knowles, T.P.; et al. Soluble aggregates present in cerebrospinal fluid change in size and mechanism of toxicity during Alzheimer’s disease progression. Acta Neuropathol. Commun. 2019, 7, 120. [Google Scholar] [CrossRef] [Green Version]
  133. Thompson, A.J.; Herling, T.W.; Kubánková, M.t.; Vyšniauskas, A.; Knowles, T.P.; Kuimova, M.K. Molecular rotors provide insights into microscopic structural changes during protein aggregation. J. Phys. Chem. B 2015, 119, 10170–10179. [Google Scholar] [CrossRef] [Green Version]
  134. Kubánková, M.; López-Duarte, I.; Bull, J.A.; Vadukul, D.M.; Serpell, L.C.; de Saint Victor, M.; Stride, E.; Kuimova, M.K. Probing supramolecular protein assembly using covalently attached fluorescent molecular rotors. Biomaterials 2017, 139, 195–201. [Google Scholar] [CrossRef]
  135. Needham, L.-M.; Weber, J.; Varela, J.A.; Fyfe, J.W.; Do, D.T.; Xu, C.K.; Tutton, L.; Cliffe, R.; Keenlyside, B.; Klenerman, D.; et al. ThX–a next-generation probe for the early detection of amyloid aggregates. Chem. Sci. 2020, 11, 4578–4583. [Google Scholar] [CrossRef] [Green Version]
  136. Babu, E.; Mareeswaran, P.M.; Sathish, V.; Singaravadivel, S.; Rajagopal, S. Sensing and inhibition of amyloid-β based on the simple luminescent aptamer–ruthenium complex system. Talanta 2015, 134, 348–353. [Google Scholar] [CrossRef]
  137. Tsukakoshi, K.; Abe, K.; Sode, K.; Ikebukuro, K. Selection of DNA aptamers that recognize α-synuclein oligomers using a competitive screening method. Anal. Chem. 2012, 84, 5542–5547. [Google Scholar] [CrossRef]
  138. Whiten, D.R.; Zuo, Y.; Calo, L.; Choi, M.L.; De, S.; Flagmeier, P.; Wirthensohn, D.C.; Kundel, F.; Ranasinghe, R.T.; Sanchez, S.E.; et al. Nanoscopic characterisation of individual endogenous protein aggregates in human neuronal cells. ChemBioChem 2018, 19, 2033–2038. [Google Scholar] [CrossRef] [PubMed]
  139. Köhler, G.; Milstein, C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975, 256, 495–497. [Google Scholar] [CrossRef] [PubMed]
  140. Morrison, S.L.; Johnson, M.J.; Herzenberg, L.A.; Oi, V.T. Chimeric human antibody molecules: Mouse antigen-binding domains with human constant region domains. Proc. Natl. Acad. Sci. USA 1984, 81, 6851–6855. [Google Scholar] [CrossRef] [Green Version]
  141. Omidfar, K.; Daneshpour, M. Advances in phage display technology for drug discovery. Expert Opin. Drug Discov. 2015, 10, 651–669. [Google Scholar] [CrossRef] [PubMed]
  142. Sormanni, P.; Aprile, F.A.; Vendruscolo, M. Third generation antibody discovery methods: In silico rational design. Chem. Soc. Rev. 2018, 47, 9137–9157. [Google Scholar] [CrossRef] [PubMed]
  143. Baran, D.; Pszolla, M.G.; Lapidoth, G.D.; Norn, C.; Dym, O.; Unger, T.; Albeck, S.; Tyka, M.D.; Fleishman, S.J. Principles for computational design of binding antibodies. Proc. Natl. Acad. Sci. USA 2017, 114, 10900–10905. [Google Scholar] [CrossRef] [Green Version]
  144. Li, T.; Pantazes, R.J.; Maranas, C.D. OptMAVEn–a new framework for the de novo design of antibody variable region models targeting specific antigen epitopes. PLoS ONE 2014, 9, e105954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Adolf-Bryfogle, J.; Kalyuzhniy, O.; Kubitz, M.; Weitzner, B.D.; Hu, X.; Adachi, Y.; Schief, W.R.; Dunbrack, R.L., Jr. RosettaAntibodyDesign (RAbD): A general framework for computational antibody design. PLoS Comput. Biol. 2018, 14, e1006112. [Google Scholar] [CrossRef] [Green Version]
  146. Aprile, F.A.; Sormanni, P.; Perni, M.; Arosio, P.; Linse, S.; Knowles, T.P.; Dobson, C.M.; Vendruscolo, M. Selective targeting of primary and secondary nucleation pathways in Aβ42 aggregation using a rational antibody scanning method. Sci. Adv. 2017, 3, e1700488. [Google Scholar] [CrossRef] [Green Version]
  147. Ehrenberg, A.J.; Khatun, A.; Coomans, E.; Betts, M.J.; Capraro, F.; Thijssen, E.H.; Senkevich, K.; Bharucha, T.; Jafarpour, M.; Young, P.N.; et al. Relevance of biomarkers across different neurodegenerative diseases. Alzheimers Res. Ther. 2020, 12, 56. [Google Scholar] [CrossRef]
  148. Klunk, W.E.; Engler, H.; Nordberg, A.; Wang, Y.; Blomqvist, G.; Holt, D.P.; Bergström, M.; Savitcheva, I.; Huang, G.F.; Estrada, S.; et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann. Neurol. Off. J. Am. Neurol. Assoc. Child Neurol. Soc. 2004, 55, 306–319. [Google Scholar] [CrossRef] [PubMed]
  149. Meier, S.R.; Syvänen, S.; Hultqvist, G.; Fang, X.T.; Roshanbin, S.; Lannfelt, L.; Neumann, U.; Sehlin, D. Antibody-based in vivo PET imaging detects amyloid-β reduction in Alzheimer transgenic mice after BACE-1 inhibition. J. Nucl. Med. 2018, 59, 1885–1891. [Google Scholar] [CrossRef] [Green Version]
  150. Nabuurs, R.J.; Rutgers, K.S.; Welling, M.M.; Metaxas, A.; De Backer, M.E.; Rotman, M.; Bacskai, B.J.; Van Buchem, M.A.; Van der Maarel, S.M.; van Der Weerd, L. In vivo detection of amyloid-β deposits using heavy chain antibody fragments in a transgenic mouse model for Alzheimer’s disease. PLoS ONE 2012, 7, e38284. [Google Scholar] [CrossRef] [Green Version]
  151. Spires-Jones, T.L.; Attems, J.; Thal, D.R. Interactions of pathological proteins in neurodegenerative diseases. Acta Neuropathol. 2017, 134, 187–205. [Google Scholar] [CrossRef] [PubMed]
  152. Colom-Cadena, M.; Gelpi, E.; Charif, S.; Belbin, O.; Blesa, R.; Martí, M.J.; Clarimon, J.; Lleó, A. Confluence of α-synuclein, tau, and β-amyloid pathologies in dementia with Lewy bodies. J. Neuropathol. Exp. Neurol. 2013, 72, 1203–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. de Wolf, F.; Ghanbari, M.; Licher, S.; McRae-McKee, K.; Gras, L.; Weverling, G.J.; Wermeling, P.; Sedaghat, S.; Ikram, M.K.; Waziry, R.; et al. Plasma tau, neurofilament light chain and amyloid-β levels and risk of dementia; a population-based cohort study. Brain 2020, 143, 1220–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Majbour, N.K.; Vaikath, N.N.; van Dijk, K.D.; Ardah, M.T.; Varghese, S.; Vesterager, L.B.; Montezinho, L.P.; Poole, S.; Safieh-Garabedian, B.; Tokuda, T.; et al. Oligomeric and phosphorylated alpha-synuclein as potential CSF biomarkers for Parkinson’s disease. Mol. Neurodegener. 2016, 11, 7. [Google Scholar] [CrossRef] [Green Version]
  155. Bourbouli, M.; Rentzos, M.; Bougea, A.; Zouvelou, V.; Constantinides, V.C.; Zaganas, I.; Evdokimidis, I.; Kapaki, E.; Paraskevas, G.P. Cerebrospinal fluid TAR DNA-binding protein 43 combined with tau proteins as a candidate biomarker for amyotrophic lateral sclerosis and frontotemporal dementia spectrum disorders. Dement. Geriatr. Cogn. Disord. 2017, 44, 144–152. [Google Scholar] [CrossRef]
  156. Appel-Cresswell, S.; Vilarino-Guell, C.; Encarnacion, M.; Sherman, H.; Yu, I.; Shah, B.; Weir, D.; Thompson, C.; Szu-Tu, C.; Trinh, J.; et al. Alpha-synuclein p. H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. 2013, 28, 811–813. [Google Scholar] [CrossRef]
  157. de Lassichère, C.C.; Mai, T.D.; Taverna, M. Antibody-free detection of amyloid beta peptides biomarkers in cerebrospinal fluid using capillary isotachophoresis coupled with mass spectrometry. J. Chromatogr. A 2019, 1601, 350–356. [Google Scholar] [CrossRef] [PubMed]
  158. DeMarco, M.L.; Nguyen, Q.; Fok, A.; Hsiung, G.-Y.R.; van der Gugten, J.G. An automated clinical mass spectrometric method for identification and quantification of variant and wild-type amyloid-β 1-40 and 1-42 peptides in CSF. Alzheimers Dement. 2020, 12, e12036. [Google Scholar] [CrossRef]
  159. Weber, D.M.; Tran, D.; Goldman, S.M.; Taylor, S.W.; Ginns, E.I.; Lagier, R.J.; Rissman, R.A.; Brewer, J.B.; Clarke, N.J. High-Throughput Mass Spectrometry Assay for Quantifying β-Amyloid 40 and 42 in Cerebrospinal Fluid. Clin. Chem. 2019, 65, 1572–1580. [Google Scholar] [CrossRef]
  160. Freedman, K.J.; Otto, L.M.; Ivanov, A.P.; Barik, A.; Oh, S.-H.; Edel, J.B. Nanopore sensing at ultra-low concentrations using single-molecule dielectrophoretic trapping. Nat. Commun. 2016, 7, 10217. [Google Scholar] [CrossRef] [Green Version]
  161. Wang, X.; Wilkinson, M.D.; Lin, X.; Ren, R.; Willison, K.R.; Ivanov, A.P.; Baum, J.; Edel, J.B. Single-molecule nanopore sensing of actin dynamics and drug binding. Chem. Sci. 2020, 11, 970–979. [Google Scholar] [CrossRef] [Green Version]
  162. Japrung, D.; Dogan, J.; Freedman, K.J.; Nadzeyka, A.; Bauerdick, S.; Albrecht, T.; Kim, M.J.; Jemth, P.; Edel, J.B. Single-molecule studies of intrinsically disordered proteins using solid-state nanopores. Anal. Chem. 2013, 85, 2449–2456. [Google Scholar] [CrossRef] [PubMed]
  163. Rosenstein, J.K.; Wanunu, M.; Merchant, C.A.; Drndic, M.; Shepard, K.L. Integrated nanopore sensing platform with sub-microsecond temporal resolution. Nat. Methods 2012, 9, 487–492. [Google Scholar] [CrossRef]
  164. Shahnawaz, M.; Tokuda, T.; Waragai, M.; Mendez, N.; Ishii, R.; Trenkwalder, C.; Mollenhauer, B.; Soto, C. Development of a biochemical diagnosis of Parkinson disease by detection of α-synuclein misfolded aggregates in cerebrospinal fluid. JAMA Neurol. 2017, 74, 163–172. [Google Scholar] [CrossRef] [PubMed]
  165. Saijo, E.; Groveman, B.R.; Kraus, A.; Metrick, M.; Orrù, C.D.; Hughson, A.G.; Caughey, B. Ultrasensitive RT-QuIC Seed Amplification Assays for Disease-Associated Tau, α-Synuclein, and Prion Aggregates. In Protein Misfolding Diseases: Methods and Protocols; Gomes, C.M., Ed.; Springer: New York, NY, USA, 2019; pp. 19–37. [Google Scholar]
  166. Abd-Elhadi, S.; Basora, M.; Vilas, D.; Tolosa, E.; Sharon, R. Total α-synuclein levels in human blood cells, CSF, and saliva determined by a lipid-ELISA. Anal. Bioanal. Chem. 2016, 408, 7669–7677. [Google Scholar] [CrossRef]
  167. Bittner, T.; Zetterberg, H.; Teunissen, C.E.; Ostlund, R.E., Jr.; Militello, M.; Andreasson, U.; Hubeek, I.; Gibson, D.; Chu, D.C.; Eichenlaub, U.; et al. Technical performance of a novel, fully automated electrochemiluminescence immunoassay for the quantitation of β-amyloid (1–42) in human cerebrospinal fluid. Alzheimers Dement. 2016, 12, 517–526. [Google Scholar] [CrossRef] [Green Version]
  168. Song, L.; Lachno, D.R.; Hanlon, D.; Shepro, A.; Jeromin, A.; Gemani, D.; Talbot, J.A.; Racke, M.M.; Dage, J.L.; Dean, R.A. A digital enzyme-linked immunosorbent assay for ultrasensitive measurement of amyloid-β 1–42 peptide in human plasma with utility for studies of Alzheimer’s disease therapeutics. Alzheimers Res. Ther. 2016, 8, 58. [Google Scholar] [CrossRef] [Green Version]
  169. Kühbach, K.; Hülsemann, M.; Herrmann, Y.; Kravchenko, K.; Kulawik, A.; Linnartz, C.; Peters, L.; Wang, K.; Willbold, J.; Willbold, D.; et al. Application of an amyloid beta oligomer standard in the sFIDA assay. Front. Neurosci. 2016, 10, 8. [Google Scholar] [CrossRef] [Green Version]
  170. Wang, L.-S.; Leung, Y.Y.; Chang, S.-K.; Leight, S.; Knapik-Czajka, M.; Baek, Y.; Shaw, L.M.; Lee, V.M.-Y.; Trojanowski, J.Q.; Clark, C.M. Comparison of xMAP and ELISA assays for detecting cerebrospinal fluid biomarkers of Alzheimer’s disease. J. Alzheimers Dis. 2012, 31, 439–445. [Google Scholar] [CrossRef] [Green Version]
  171. Singer, D.; Soininen, H.; Alafuzoff, I.; Hoffmann, R. Immuno-PCR-based quantification of multiple phosphorylated tau-epitopes linked to Alzheimer’s disease. Anal. Bioanal. Chem. 2009, 395, 2263–2267. [Google Scholar] [CrossRef] [PubMed]
  172. Stegurová, L.; Dráberová, E.; Bartos, A.; Dráber, P.; Řípová, D.; Dráber, P. Gold nanoparticle-based immuno-PCR for detection of tau protein in cerebrospinal fluid. J. Immunol. Methods 2014, 406, 137–142. [Google Scholar] [CrossRef] [PubMed]
  173. Hashimoto, M.; Aoki, M.; Winblad, B.; Tjernberg, L.O. A novel approach for Aβ1–40 quantification using immuno-PCR. J. Neurosci. Methods 2012, 205, 364–367. [Google Scholar] [CrossRef] [PubMed]
  174. Wei, T.-Y.; Fu, Y.; Chang, K.-H.; Lin, K.-J.; Lu, Y.-J.; Cheng, C.-M. Point-of-care devices using disease biomarkers to diagnose neurodegenerative disorders. Trends Biotechnol. 2018, 36, 290–303. [Google Scholar] [CrossRef]
  175. Shao, Y.; Le, W. Recent advances and perspectives of metabolomics-based investigations in Parkinson’s disease. Mol. Neurodegener. 2019, 14, 3. [Google Scholar] [CrossRef] [Green Version]
  176. Phelan, M.; Caamaño-Gutiérrez, E.; Gant, M.; Grosman, R.; Madine, J. Using an NMR metabolomics approach to investigate the pathogenicity of amyloid-beta and alpha-synuclein. Metabolomics 2017, 13, 151. [Google Scholar] [CrossRef] [Green Version]
  177. Zhang, Y.; Liu, Z.; Ji, B.; Liu, L.; Wu, S.; Liu, X.; Wang, S.; Wang, L. Metabolite Profile of Alzheimer’s Disease in the Frontal Cortex as Analyzed by HRMAS 1H NMR. Front. Aging Neurosci. 2019, 10, 424. [Google Scholar] [CrossRef]
  178. Zhu, S.; Wuolikainen, A.; Wu, J.; Öhman, A.; Wingsle, G.; Moritz, T.; Andersen, P.M.; Forsgren, L.; Trupp, M. Targeted multiple reaction monitoring analysis of CSF identifies UCHL1 and GPNMB as candidate biomarkers for ALS. J. Mol. Neurosci. 2019, 69, 643–657. [Google Scholar] [CrossRef] [Green Version]
  179. Hilton, D.; Stephens, M.; Kirk, L.; Edwards, P.; Potter, R.; Zajicek, J.; Broughton, E.; Hagan, H.; Carroll, C. Accumulation of α-synuclein in the bowel of patients in the pre-clinical phase of Parkinson’s disease. Acta Neuropathol. 2014, 127, 235–241. [Google Scholar] [CrossRef]
  180. Gries, M.; Christmann, A.; Schulte, S.; Weyland, M.; Rommel, S.; Martin, M.; Baller, M.; Röth, R.; Schmitteckert, S.; Unger, M.; et al. Functional and molecular early enteric biomarkers for Parkinson’s disease in mice and men. bioRxiv 2020. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of amyloid aggregation. In normal conditions, proteins are newly synthesized on ribosomes and released to play biological functions. In stress conditions or as a consequence of pathological post-translational modifications (PTMs), these proteins can assemble into small soluble oligomers and ultimately form insoluble amyloid fibrils. PTMs may occur at any state of this process, affecting the formation, stability and toxicity of these aggregates.
Figure 1. Schematic representation of amyloid aggregation. In normal conditions, proteins are newly synthesized on ribosomes and released to play biological functions. In stress conditions or as a consequence of pathological post-translational modifications (PTMs), these proteins can assemble into small soluble oligomers and ultimately form insoluble amyloid fibrils. PTMs may occur at any state of this process, affecting the formation, stability and toxicity of these aggregates.
Ijms 22 04128 g001
Figure 2. Aggregated protein species and representative probes used for their detection.
Figure 2. Aggregated protein species and representative probes used for their detection.
Ijms 22 04128 g002
Figure 3. Detection of amyloidogenic proteins at different stages of protein aggregation. Main representative strategies and techniques are shown.
Figure 3. Detection of amyloidogenic proteins at different stages of protein aggregation. Main representative strategies and techniques are shown.
Ijms 22 04128 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jin, Y.; Vadukul, D.M.; Gialama, D.; Ge, Y.; Thrush, R.; White, J.T.; Aprile, F.A. The Diagnostic Potential of Amyloidogenic Proteins. Int. J. Mol. Sci. 2021, 22, 4128. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084128

AMA Style

Jin Y, Vadukul DM, Gialama D, Ge Y, Thrush R, White JT, Aprile FA. The Diagnostic Potential of Amyloidogenic Proteins. International Journal of Molecular Sciences. 2021; 22(8):4128. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084128

Chicago/Turabian Style

Jin, Yiyun, Devkee Mahesh Vadukul, Dimitra Gialama, Ying Ge, Rebecca Thrush, Joe Thomas White, and Francesco Antonio Aprile. 2021. "The Diagnostic Potential of Amyloidogenic Proteins" International Journal of Molecular Sciences 22, no. 8: 4128. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084128

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

Article Metrics

Back to TopTop