Abstract
Synapses are the site for brain communication where information is transmitted between neurons and stored for memory formation. Synaptic degeneration is a global and early pathogenic event in neurodegenerative disorders with reduced levels of pre- and postsynaptic proteins being recognized as a core feature of Alzheimer’s disease (AD) pathophysiology. Together with AD, other neurodegenerative and neurodevelopmental disorders show altered synaptic homeostasis as an important pathogenic event, and due to that, they are commonly referred to as synaptopathies. The exact mechanisms of synapse dysfunction in the different diseases are not well understood and their study would help understanding the pathogenic role of synaptic degeneration, as well as differences and commonalities among them and highlight candidate synaptic biomarkers for specific disorders. The assessment of synaptic proteins in cerebrospinal fluid (CSF), which can reflect synaptic dysfunction in patients with cognitive disorders, is a keen area of interest. Substantial research efforts are now directed toward the investigation of CSF synaptic pathology to improve the diagnosis of neurodegenerative disorders at an early stage as well as to monitor clinical progression. In this review, we will first summarize the pathological events that lead to synapse loss and then discuss the available data on established (eg, neurogranin, SNAP-25, synaptotagmin-1, GAP-43, and α-syn) and emerging (eg, synaptic vesicle glycoprotein 2A and neuronal pentraxins) CSF biomarkers for synapse dysfunction, while highlighting possible utilities, disease specificity, and technical challenges for their detection.
Keywords: Synaptic biomarkers, cerebrospinal fluid, synaptopathies, Alzheimer’s disease, proteomics
Introduction
The central nervous system (CNS) can be subject to numerous pathological conditions, which can affect its development, functionality, or cause premature cell death, resulting in neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Although these disorders have different etiologies and pathophysiological mechanisms, many of them have some degree of dysfunction and alteration of the synapses and can thus be categorized as synaptopathies.1,2 In this review, we will discuss how synapses are affected in the most common diseases affecting the CNS, and how advances in synaptic biomarker discovery provide new tools for the study of those diseases. We will mainly focus on neurodegenerative conditions, and in particular on Alzheimer’s disease (AD), which is the predominant cause of dementia affecting approximately 50 million people worldwide.3 Although the exact mechanisms of synaptic loss and dysfunction in the different diseases are still poorly understood, there is evidence that a reduction in synaptic activity and density is one of the earliest events in many of the diseases of the CNS and may even appear before neuronal loss.4,5 The significant role of synapse dysfunction in the disease pathology and progression of synaptopathies has therefore prompted a keen interest in detecting and quantifying synaptic proteins. Molecular brain imaging6 and analysis of cerebrospinal fluid (CSF)7 are used in conjunction to study synaptic proteins, with the aim of using them as biomarkers for prognosis, to follow disease progression and to evaluate effects of drug testing.
Pathophysiology of Synaptic Dysfunction and Loss
Synaptic functions
The neuronal synapses are the functional units of neurotransmission in the brain, with an estimated 100 trillion interconnecting synapses8 in an elaborate and complex network. Synapses are formed during development and the early postnatal period. After reaching the maximum density at 2 to 4 years of age,9,10 in the following years synapses are physiologically eliminated in a process known as pruning.11 Synapses that survive to adulthood are the ones stably maintained, although we have a certain degree of synapse formation and elimination throughout life.12
Neuronal signal transmission in the CNS requires the presence of functional synapses, with properly arranged pre- and postsynaptic compartments. The presynaptic compartment contains all the structures for formation, storage, and release of neurotransmitter-containing vesicles. Following an action potential, the increase of Ca2+ in the presynaptic terminal triggers synaptic vesicles to fuse with the presynaptic membrane upon which neurotransmitters are released into the synaptic cleft.13 Subsequently, neurotransmitters interact with receptors on the postsynaptic compartment (the dendritic spine), and through the activation of different signaling pathways14 the neuronal signal is transmitted further. Synapses can be excitatory or inhibitory, using glutamate and GABA, as neurotransmitters, respectively.15 The dendritic spines are the primary location of excitatory synapses.
Synapse formation, maturation, and elimination is a dynamic series of events that can be defined as synaptic plasticity. Processes representing synaptic plasticity are phenomena termed long-term potentiation (LTP) and long-term depression (LTD), through which, during memory formation, signaling via preferred synapses is enhanced or reduced. Selection of synapses seems to be activity-dependent, LTP is usually considered as a protective mechanism and LTD as inductive of elimination.16 These 2 processes are considered the basis for memory formation and storage.17,18 LTP is identified by the addition of new receptors at the postsynaptic density (PSD) and the consequent enlargement of the spine head resulting in transmission of a stronger signal.19 On the contrary, during LTD a series of events lead to spine shrinkage and elimination.18 Many different mechanisms for synaptic elimination have been suggested (for extensive review, see Cardozo et al20 and Maiti et al21) Elimination of weaker and unnecessary synapses and maintenance of the stronger ones are processes that balance each other, to ensure proper connectivity between brain regions and signal refinement.22,23 For proper synaptic activity, a balance is needed, and alterations between synapse formation and elimination can cause synaptic dysfunction and impaired brain network activities.21 To understand pathological mechanisms and at which stage synapses are affected is of utmost importance to define targets and intervention strategies.
Synapse and neuronal loss in brain disorders
As mentioned, a balance in synapse formation and pruning is essential for proper connectivity and brain functionality. For instance, excessive synaptic pruning during adolescence is one of the hypothesized mechanisms for schizophrenia, which most commonly manifests with an onset in late adolescence or early-adulthood.24-26 The term “synaptopathy” is applied to refer to all diseases that are characterized by a progressive synaptic dysfunction and loss.20 AD, the most common neurodegenerative disease, can be therefore considered both a synaptopathy and a proteinopathy.
AD pathology is identified by the presence of extracellular deposits of amyloid-β (Aβ) plaques, formed by the aggregation of Aβ peptides, and neurofibrillary tangles (NFTs) that are intraneuronal accumulations of hyperphosphorylated and truncated tau protein, respectively.27,28 Along with these main hallmarks, gliosis, neuroinflammation,29-31 and vascular dysfunction32,33 are also present, which reflects the complexity of AD. However, it is synaptic loss which best correlates with cognitive symptoms34-36 and it is also apparent in the early stages of the disease pathophysiology.37,38 The number of synapses in the brain decreases during normal aging but this decrease is exacerbated in AD and, consequently, the synapse-to-neuron ratio is lower in AD brains compared with age-matched non-demented individuals.39 In AD, brain biopsies show synaptic loss in neocortical regions and the hippocampus,40,41 the latter showing the greatest reduction by approximately 50%.42-44
How the major AD hallmarks, tau and Aβ, pathologically interact with synapses needs more investigation. However, most studies identified the oligomeric forms of Aβ and tau, rather than larger aggregates, to be the synaptotoxic species.45-49 Both in vivo50 and ex vivo51 Aβ oligomers (Aβo) disrupt LTP, probably interfering with NMDAR (N-methyl-d-aspartate receptor) activity and downstream pathways,52 in addition to causing oxidative stress, impairing axonal transport, and causing nerve cell death (reviewed in Cline et al53). In AD, Aβ and tau act in concert and studies have identified their simultaneous presence in the postsynaptic compartment.54,55 Aβo have been suggested to bind to a variety of targets,56 including cellular prion protein (PrPc),57 neuroligin 1, neurexin-2α,58 PirB, EphB2,59 shank, synGap, Na/K-ATPase,60 ultimately leading to impairment of LTP and synapse loss.45,50,61 Phosphorylated tau oligomers can relocate from axons to dendrites, interfering with NMDAR and AMPAR (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor) and impairing glutamatergic transmission.62,63
Moreover, as points of transmission of signals between neurons, synapses seem also to help the spreading of the pathology via prion-like mechanisms, and some studies show the possibility of Aβo and tau oligomers to be transferred from neuron to neuron.64-69
Tau aggregation, without Aβ pathology, is also a pathological hallmark of other neurodegenerative diseases, the so-called tauopathies.70 Tauopathies include, among others, some forms of frontotemporal lobar degeneration (FTLD), namely FTLD-tau, progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD). Although all tauopathies have in common the presence of tau aggregates in the CNS, the characteristics of these aggregates differ among them and are different from the NFTs of AD. PSP shows filamentous aggregates in astrocytes and oligodendrocytes, while in CBD tau accumulation in neurons is less fibrillar and in astrocytes it accumulates in the form of astrocytic plaques.71 As introduced above, oligomeric tau has been connected to synaptic damage through different pathways,63 also involving activation of microglia and astrocytes through inflammatory processes,72 and animal models of tau pathology show early synaptic loss prior to neuronal death.73,74
As the combination of Aβ and tau pathology define AD, accumulation of aggregated α-synuclein (α-syn) is the pathological feature of several diseases, which are commonly collectively referred to as α-synucleinopathies.75 Among the most common synucleopathies are Parkinson’s disease (PD), Parkinson’s disease dementia (PDD), dementia with Lewy body (DLB), and multiple system atrophy (MSA). PD, PDD, and DLB show accumulation of the so-called Lewy neurites (LN) and Lewy bodies (LB), where α-syn is the principal component.76 The physiological role of α-syn at the synapse is not precisely understood yet77; however, it is commonly accepted that its dyshomeostasis and accumulation leads to cell damage and it is responsible for synaptic impairment and neuronal damage.78,79 Alpha-syn, localized mainly presynaptically, is involved in synaptic vesicle regulation and trafficking,80,81 and in the SNARE complex formation.82,83 It also interacts with membrane lipids and can associate with mitochondria,84 the Golgi-endoplasmic reticulum system,85 and the endolysosomal system. Pathologic α-syn can interfere with all these organelles, consequently impairing-related pathways.86,87 The protein can change its conformation and aggregate, giving rise to oligomers, fibrils, and larger aggregates.88 Which form is responsible for toxicity is still a matter of debate. However, as discussed for AD, the oligomeric form of α-syn has been suggested to be the responsible for the synaptic damage in dopaminergic neurons,89,90 and the possibility for oligomeric α-syn of spreading in a prion-like manner has been proposed.91,92 At a cellular level, typically PD and DLB are distinguished from MSA, inasmuch the accumulations of LN and LB are mainly present in neurons, while in MSA, α-syn accumulation appears in oligodendrocytes.93 Moreover, MSA inclusions seem to be more compact and aggressive,94 in line with the increased severity of the disease.95 However, it was most recently reported that also neurons in MSA show α-syn oligomers depositions.96 A recent study showed that α-syn filaments differ in DLB and MSA.97 PD and DLB are usually distinguished based on the symptoms, with DLB being the second most common type of dementia.98 PD, similar to AD, starts many years before symptoms become overt and at that point, patients had already lost up to 60% motor neurons in the substantia nigra.99
Nonetheless, α-syn was first identified and characterized in relation to AD when it was found to be a major non-amyloid beta component of Aβ plaques.100 In fact, Lewy pathology can be also found in over half of all patients with AD.101-103 To further complicate the clinical picture, α-syn depositions are found in tauopathies like PSP and CBD, and NFTs have been found in PD brains.104,105
Comorbidities and co-occurrence of different pathologies make diagnosis of these diseases challenging. Synapse damage is a common and early first change in the disease development and prolonged synaptic damage can lead to synaptic loss. Neuronal damage and death seem to be a follow-up event seen only at later stages. For these reasons, the investigation of synaptic biomarkers has the potential to find a way to diagnose the disease in its early stages and also to give us information on the main pathological mechanisms involved.
Current Climate of Fluid Biomarkers in Dementia
Imaging and CSF biomarkers
The core CSF biomarkers for AD (Aβ42/Aβ40, total-tau, and phospho-tau), reflecting the defining Aβ and tau neuropathologies, consistently demonstrate diagnostically significant changes across studies106 and now have prominent positions in biological and diagnostic criteria for AD.28,107 The concentrations of these core AD biomarkers, however, are no different from healthy controls in the majority of dementias outside of the AD continuum108,109 which can be of great utility in the differential diagnosis of patients with cognitive symptoms. An exception can be made for Creutzfeldt–Jakob disease (CJD), which presents vastly increased levels of t-tau, whereas the concentrations of p-tau181 remain normal or only marginally changed in CJD.110,111
Together with CSF biomarkers, positron emission tomography (PET) and magnetic resonance imaging (MRI) are used to provide a clearer view of pathology and atrophy patterns in the brains of living humans. MRI allows for the measurement of brain atrophy and provides information on regional, structural, and functional integrity of the brain.112 In the research of neurodegenerative disorders, PET tracers for protein aggregation such as Aβ113,114 and tau,115 as well as glucose metabolism as a measure for neuronal activity116,117 and synaptic density,118 have been developed. Together with CSF biomarkers, MRI and PET are nowadays included in the research diagnostic criteria for AD.28,119 However, the availability of PET scans is limited and when possible expensive, thus it is not always applicable.
Blood biomarkers
In certain instances, the biomarkers field is rapidly evolving from CSF into blood, which is a more easily accessible biological fluid. Despite the latest advancement in developing CSF biomarkers for synaptic integrity and large high-resolution mass spectrometry proteomic studies demonstrating the presence of synaptic proteins in blood,120,121 to date no studies have shown positive results for any pre- or postsynaptic biomarkers in blood correlating to any neurodegenerative disease phenotype.
The advancement of ultrasensitive methodologies has enabled, however, the detection of the CSF core biomarkers and neuronal injury, like neurofilaments, in blood. New evidence from high-resolution mass spectrometry,122,123 single molecule array (Simoa),124 and fully automated immunoassays,125 which are highly sensitive and alleviate confounding matrix effects in blood, suggests that Aβ peptide ratios are specific markers of individuals with Aβ- positive brain scans. In addition, recent evidence has shown that plasma p-tau181 concentrations are higher in individuals with AD dementia than in healthy controls.126 Plasma p-tau181 correlates with tau PET in Aβ-positive AD individuals and, encouragingly, can accurately identify elderly controls and mild cognitive impairment (MCI) individuals with a positive Aβ-PET scan (area under the curve [AUC] > 0.85).127-129 Conversely, although significant increase of plasma t-tau has been vastly observed in individuals with AD, the plasma t-tau levels between control, MCI, and AD groups substantially overlapped.130
The neurofilaments are cytoskeletal protein abundantly expressed in neuronal axons, among which neurofilament light polypeptide (NfL) is the smallest of the neurofilament proteins (for a detailed review on neurofilament structure and function, please see Khalil et al131). A moderate-to-good correlation between NfL concentration in blood and CSF has been observed in several studies and many CSF findings of increased NfL in neurodegenerative diseases have subsequently been replicated in blood.132 Although not a specific marker for AD, blood NfL has the potential to track or predict many aspects of neurodegeneration, including cognitive performance,133 the degree of postmortem pathology,134 structural imaging,135 and glucose metabolism.136,137
Fluid Biomarkers for Synapse Pathology
The pathophysiology of synaptopathies and the significance of synapses in cognition make a convincing argument for the need and use of biomarkers of synapse pathology as representation of cognitive and synaptic function. Clinically, synaptic biomarkers may link synaptic degeneration with the cognitive status and decline of the patient, and they could be implemented together with cognitive tests to have a more precise description of the patient’s symptoms, especially at early stages. Moreover, synaptic biomarkers can help to understand the underlying pathological processes ongoing during cognitive diseases, as different proteins could reflect different mechanisms, thus helping the diagnosis. In addition, synaptic biomarkers can also be used during drug development, to monitor the efficacy of treatments on synaptic functioning in drug trials.
Pre- and postsynaptic biomarkers
Biomarkers for synaptic dysfunction can be divided into pre- and postsynaptic biomarkers depending on the localization of the protein. The presence of synaptic proteins in CSF was first demonstrated in the late 1990s,138,139 but for a long time, most studies still involved postmortem brain tissue. However, in the last decade, advances in mass spectrometry and immunoassays have allowed the accurate quantification of synaptic proteins in biofluids. As of today, there are 4 main presynaptic biomarkers, growth-associated protein 43 (GAP-43), synaptosomal-associated protein 25 (SNAP-25), synaptotagmin-1, and α-syn, and 1 postsynaptic marker, neurogranin.
GAP-43
GAP-43 is a presynaptic protein which plays an important role in memory and information storage.140 It is anchored on the cytoplasmic side of the presynaptic plasma membrane and is mainly expressed in the hippocampus, entorhinal cortex, and neocortex of the adult brain. At the synapse, upon intracellular Ca2+ increase, GAP-43 is phosphorylated by protein kinase C. This leads GAP-43 to interact, among others, with synaptophysin and SNAP-25, facilitating synaptic vesicle recycling.141 Studies have found GAP-43 CSF levels to be significantly increased in patients with AD compared with controls142 and also other neurodegenerative disorders.143 CSF GAP-43 levels were also increased in preclinical and clinical patients with AD compared with controls. However, in an antibody-based explorative study, no significant changes in patients with PD or DLB were found in comparison with controls.144 Altered CSF GAP-43 levels have also been reported in progressive multiple sclerosis (MS),143,145 inflammation,146 stroke,147 and PD,109 but not in frontotemporal dementia (FTD).109
SNAP-25
SNAP-25 is a presynaptic protein with a key role in neuronal survival and cognitive function due to its essential part in vesicular exocytosis, neurite outgrowth, and LTP.148 SNAP-25, together with vesicle-associated membrane proteins (VAMPs) and syntaxins, forms SNARE complexes, which mediate synaptic vesicle apposition to the presynaptic membrane thus allowing for the Ca2+-triggered vesicle fusion during exocytosis.149 SNAP-25 has, in various studies using both enzyme-linked immunosorbent assay (ELISA) and mass spectrometry-based assays, shown to have significantly higher CSF levels in AD, even at a very early stages.7,150-152 Increased CSF levels of SNAP-25 have also been found in patients with PD153 and patients with sporadic CJD.154 In addition, SNAP-25 has been associated with several psychiatric diseases such as attention deficiency hyperactivity disorder (ADHD), schizophrenia, and bipolar disorder.149 Furthermore, there are 2 splicing variants of SNAP-25: SNAP-25A and SNAP-25B. Mass spectrometry–based methods to quantify both total SNAP-25 and the 2 isoforms have therefore been developed to study potential differences in the roles of the isoforms of SNAP-25 in disease. Nine amino acids differentiate the 2 protein isoforms, which also differ in their effects on neurotransmission.155 To our knowledge, no studies have investigated the different isoforms in CSF. However, a postmortem brain tissue study by Barakauskas et al155 found significantly decreased levels of total SNAP-25 and SNAP-25A but not of SNAP-25B, indicating a specific differential expression of SNAP-25A in schizophrenia.
Synaptotagmin-1
Synaptotagmin-1 is a calcium sensor vesicle protein vital for fast synchronous neurotransmitter release in hippocampal neurons.156 It is a transmembrane protein anchored in the vesicle membranes containing 2 Ca2+-binding domains. In response to Ca2+-binding at elevated concentrations, synaptotagmin-1 triggers the vesicle fusion, but the exact molecular mechanisms remain to be elucidated (for review see Park and Ryu157). Initial CSF studies of synaptotagmin-1 found it to be decreased in a CSF pool from patients with early-onset AD compared with a CSF pool from healthy controls.138 Two decades later, Öhrfelt et al158 quantified synaptotagmin-1 in individual CSF samples from patients with AD, MCI, and controls, demonstrating significantly increased concentrations of synaptotagmin-1 in patients with AD and MCI, the highest being MCI due to AD. These findings have been corroborated in a recent study where synaptotagmin-1 was quantified in patients in the AD continuum and cognitive decline from other dementias.159,160
In the same study by Tible et al, in addition to synaptotagmin-1, the concentrations of GAP-43 and SNAP-25 were quantified. All 3 presynaptic proteins were significantly increased in AD and MCI-AD compared with the other disorders. However, only SNAP-25 and GAP-43 levels were also significantly higher in AD versus MCI-AD. Only synaptotagmin-1 concentrations were significantly lower in other neurodegenerative disorders compared with controls. Recently, Clarke et al161 compared synaptotagmin-1 and SNAP-25 concentrations in patients with FTD and demonstrated increased levels in patients with AD biomarker profile compared with those patients with an FTD profile.
Alpha-synuclein and its forms
Alpha-syn is a key player in the etiology of different neurodegenerative conditions and as such, it has been studied as a possible biomarker for their detection. However, besides being a possible cause for diseases, it is also a presynaptic protein, taking part in many synaptic processes as previously described, which is why it is important to include it in this review. The synucleins family comprises α-, β-, and γ-synuclein, which are soluble proteins encoded by 3 different genes. Among them, α-syn is the most studied.162
Total α-syn
Studies of α-syn in CSF have mainly been focused on α-synucleinopathies and were based on immunological assays measuring total α-syn (t-α-syn); however, they have been largely inconsistent. For instance, in PD compared with controls, t-α-syn has been found in several studies to be slightly decreased163-165 which is supported by several meta-analyses which concluded that there are significantly lower levels of t-α-syn in PD (10%-15%). However, in other studies no significant difference has been found,166,167 and the diagnostic performance of t-α-syn is not considered sufficient for clinical utility due to significant overlap between the populations.168-170 Other synucleinopathies, like DLB and MSA, have also shown a similar decrease compared with healthy controls, while tauopathies such as PSP and CBD seem to show no significant difference.163,165,171 For AD in comparison with healthy controls, t-α-syn levels seem to be elevated171-174; however, several studies showed no significant difference.175-179 Patients with CJD, on the other hand, have a more pronounced increase in CSF t-α-syn, both compared with controls and with other neurodegenerative diseases.178,180,181 An explanation for the inconclusive findings of CSF α-syn is that leakage into the CSF from synapse breakdown occurs simultaneously as α-syn is retained in pathological inclusions. In addition, the extensive reduction in synapse number over time might lead to a decrease of α-syn production. Together these events might contribute to the confounding results.166,182 Another contributing factor for the varying results might be due to technical variation such as handling of samples or quantification methods leading to low reproducibility.182 Moreover, α-syn is largely expressed outside of the CNS and highly abundant in blood, with red blood cells (RBCs) as its major source. Thus, blood contamination during CSF acquisition might represent another source of variation, skewing the t-α-syn concentration results in CSF.183,184
Despite the possible problems just discussed, there have been many studies evaluating α-syn as blood biomarker for dementias. Studies for α-syn in plasma and serum in PD have all shown similar conflicting results as in CSF.185-188 However, a meta-analysis indicates that plasma t-α-syn is significantly higher in PD than in controls.189 In a study by Laske et al,190 decreased serum concentrations of t-α-syn in DLB were found but with no difference for AD compared with controls. There are also a few studies on RBC t-α-syn191-193 which showed significantly decreased levels of the protein in PD and AD compared with controls. Studies on t-α-syn in saliva have also been performed, but with limited success in differentiating PD from controls.194-196
Oligomeric, phosphorylated, and aggregated forms of α-synuclein
The inconclusive results of t-α-syn as a diagnostic biomarker have sparked research into pathological forms of α-syn, such as oligomeric (o-α-syn), phosphorylated (Ser129) (p-α-syn), and aggregated forms of α-synuclein. Oligomeric α-syn in CSF seems to be increased in PD compared with controls169 but not in AD and DLB.174,197 Furthermore, Parnetti et al198 found that the diagnostic accuracy of PD can be improved by using the ratio of oligomeric/total α-syn in CSF. In plasma,199 serum,200 and RBC,193,201 significantly elevated levels have been reported for PD, but also non-significant results exist.202-204 In a study by Vivacqua et al,205 increased saliva levels of o-α-syn were found for PD. Phosphorylated α-syn, one of the main disease-associated posttranslational modifications (PTMs),206 is hard to quantify due to its low CSF concentration, but similar to the oligomeric and the total form, it has been found elevated in PD169 and its diagnostic accuracy increases when its ratio to other α-syn forms are used.207 Phosphorylated-α-syn has also been indicated to be elevated in CJD181 and not increased in AD.174,179 Plasma p-α-syn has been found to be significantly increased in PD compared with controls.185,203 For the measurement of pathogenic α-syn aggregates in CSF, aggregation assays have been developed. Assays based on real-time quaking-induced conversion (RT-QuIC) or protein misfolding cyclic amplification (PMCA) have shown very promising results (specificity > 95%, sensitivity > 80%) in discriminating synucleinopathies (PD, MSA, and DLB) from nonsynucleinopathies (AD and controls).208-210
Neurogranin
Neurogranin is an intracellular 7.5-kDa protein, concentrated in the dendritic and postsynaptic compartment of synaptic spines of neurons.211,212 There it binds via its central IQ domain213 to the Ca2+-signaling mediator calmodulin, enhancing signaling for processes important in memory formation and to phosphatidic acid at the inner plasma membrane.214 A neurogranin knockout mouse model showed deficits in spatial memory and synaptic plasticity.215 In a first study, CSF neurogranin was shown by immunoprecipitation and Western blot to be increased in AD.216 After the development of immunoassay methods using ELISA,217 Singulex,218 and Mesoscale,219 these findings have been verified in several studies and neurogranin consistently showed increased levels in CSF of AD patients as compared with controls.121,220-224 This increase appears to be specific for AD, as CSF from patients with other neurodegenerative diseases, with the exception of CJD,225 do not show such an increase.161,226,227 High levels of neurogranin in CSF during prodromal AD have been shown to be predictive of more rapid progression toward AD.217,219
Besides full-length neurogranin, CSF contains mainly fragments of the C-terminal half (with a variety of different truncations at their C-terminal and N-terminal ends).216,217 Two intracellular enzymes have been identified that can generate cleavages in the functionally important IQ domain and at the very C-terminal end (calpain-1 and prolyl endopeptidase, respectively).228 Whether these different fragments of neurogranin have roles in different physiological or pathophysiological functions is still unknown. In a comparison study, different ELISAs and the Singulex assay were found to have similar performance in predicting AD, in spite targeting different parts of neurogranin.229 However, this does not rule out that particular neurogranin fragments could yield more discriminatory power to detect AD. Overall, it can be said that neurogranin may be a useful biomarker in CSF to detect early degeneration of neurons and it appears to be fairly specific for AD among several tauopathies.
Plasma concentrations of neurogranin are detectable with conventional ELISAs but are unchanged in AD and do not correlate with CSF neurogranin, probably due to the contribution of peripherally expressed neurogranin peptides to blood neurogranin measurements.121,220
Emerging synaptic biomarkers
Recently, other studies identified more synaptic proteins in CSF, which have been investigated without success so far or that show promise as synaptic biomarkers, thus worth to be mentioned in this section. Wesenhagen et al have recently reviewed 29 proteomic studies that investigated AD-related changes in CSF protein abundances. In total, 97 proteins, including the synaptic proteins neurofascin, NPTX1, NPTX2, and neurexin 1, were reported by 2 or more studies and associated with AD.230 One of the reviewed studies231 reported a synaptic biomarker panel where only the 3 synaptic proteins neurofascin, NPTX1, and neurexin 1 were significantly lowered in AD. Similarly, Lleó et al232 found that 6 synaptic proteins, calsyntenin-1, glutamate receptor 4 (GRIA4), neurexin-2A, neurexin-3A, syntaxin-1B, and thy-1 membrane glycoprotein, were increased in CSF in preclinical AD even before the core CSF biomarkers for neurodegeneration.
In explorative proteomics, high-resolution separation methods such as gel electrophoresis, isoelectric focusing, and high-performance liquid chromatography are used in conjunction with mass spectrometry and bioinformatics to study differences in protein expression due to diseases, genetic variations, or therapy. A major advantage of using an explorative approach to study protein abundances is that many hundred proteins and protein variants can be studied simultaneously without existing hypotheses or bias. Thus, the discovery of novel biomarkers could lead to new insights on disease mechanisms and eventually the formulation of novel hypotheses. However, using the explorative approach to identify biomarkers in biofluids from individual patient samples is challenging and the overlap of identified biomarker candidates among these studies has historically been relatively low. These discrepancies may be due to a low number of study participants, differences in sample handling, and other analytical parameters. Another possible approach to identify new candidate biomarkers is setting up targeted assays based on proteins of interest from studying the literature and/or public databases. Commonly shotgun proteomics, to identify possible proteins of interest, is also used in the selective process. In this way, several potential biomarker candidates can be validated in a targeted setting in larger cohorts. Among emerging synaptic biomarkers, of special note are neuronal pentraxins and the synaptic vesicle glycoprotein 2A.
Neuronal pentraxins
Neuronal pentraxin I (NPTX1, also called NP1) and II (NPTX2, also called NP2), and the neuronal pentraxin receptor (NPTXR) are widely expressed at excitatory synapses, where they bind to AMPA receptors and are suggested to be involved in synaptic plasticity.233,234 All 3 neuronal pentraxins have lately received much attention and have been shown in several studies to have decreased CSF levels in AD and MCI groups compared with controls.231,235-242 CSF pentraxin levels also correlate with cognitive performance and hippocampal volume.150,242,243 Few studies have been performed on other diseases but NPTXR has also been associated with other neurological diseases such as MS244 and FTD.245 Furthermore, in a study by Magdalinou et al246 both NPTXR and NPTX1 were found to be decreased in between atypical parkinsonian disorders (PSP, MSA, CBD) and controls.
SV2A
Recent studies using [11C]UCB-J PET have identified SV2A as the first in vivo marker of synaptic density247 which demonstrates widespread synaptic loss in AD.118 SV2A is a synaptic vesicle transmembrane protein, which in brain is widely expressed in neurons.248 SV2A has been described to be located in the dense-core vesicles249,250 and in small synaptic vesicles,251 most probably in both. Although its exact mechanism needs more investigation, it is involved in regulation of neurotransmitter release252,253 and expression and trafficking of synaptotagmin.254 Compared with the typical pattern of hypometabolism seen in AD using [18F]FDG, the spatial extent of decreases in [11C]UCB-J uptake was significantly more confined. The reduction in hippocampal binding is in line with the early loss of entorhinal cortical cell projections to the hippocampus, and reductions of hippocampal SV2A seen in postmortem studies in AD brain tissue.255,256 More recently, changes in [11C]UCB-J PET have been observed in PD,257 PSP,258 cortical basal syndrome, and epilepsy247 suggesting that SV2A could be a global marker for synaptic density, unlike CSF synaptotagmin-1, SNAP-25, GAP-43, and neurogranin, which are rather specific to AD or amyloidopathies. Recently, SV2A has been detected in CSF and shown to be reduced in AD259; however it is yet to be determined whether CSF SV2A can be used as a marker for synaptic density in other dementias and whether it has a meaningful correlation with [11C]UCB-J (Figure 1).
Figure 1.
Synaptic and neuronal biomarkers location. The picture is a schematic representation of the most studied synaptic biomarkers described in this review. As it can be noticed, most of the candidate biomarkers are localized presynaptically, with the exception of neurogranin and neuronal pentraxins (NPTX), which has also been described to be present presynaptically.260 Many proteins are involved in synaptic vesicle assembly and neurotransmitters release, like synaptotagmin-1 (syt 1), synaptophysin, SNAP-25, and SV2A.248 α-Synuclein (α-syn) can be found as a soluble form in the cytoplasm, but also associating with membrane lipids as, for instance, with synaptic vesicles and mitochondria.87 GAP-43 shows high density in the presynaptic terminal, where depending on its phosphorylation status, participates in neuronal growth modulating actin or in synaptic plasticity modulating synaptic vesicle trafficking.141 Together with actin filaments and microtubules, neurofilaments are cytoskeletal elements of the neurons, providing mechanical strength and stability.131 Tau protein, mainly expressed in axons, binds to tubulin and induce its polymerization into microtubules, which support axon outgrowth and elongation.261 α-syn indicates synuclein; DCV, dense-core vesicles; GAP-43, growth-associated protein 43; LTP, long-term potentiation; NPTX, neuronal pentraxin; SNAP-25, synaptosomal-associated protein 25.
Figure made with www.biorender.com.
Miscellaneous: other emerging synaptic biomarkers
The Rab family are key synaptic proteins involved in both recycling of neurotransmitter receptors and exocytosis of neurotransmitters. Of special note is the family member ras-related protein 3a (Rab3a), highly abundant in brain tissues, which has been connected with several neurodegenerative diseases (AD, PD, and DLB) due to its regulation of Aβ production and interaction with α-syn.262 The protein has been investigated by Bereczki et al,153 which however did not find any significant difference in CSF between patients with PD and control. A second important protein family for neurotransmitter exocytosis is the granin family, which is constituted of dense-core vesicle proteins involved, inter alia, in neuropeptide biogenesis and secretion. The proteins have not only been associated with neurodegenerative diseases, such as AD, but also with other synaptopathies, such as schizophrenia and depression.263 Three of the key granins: chromogranin-A, secretogranin-2, and neurosecretory protein VGF, have been found to have significantly lower CSF concentrations in AD.231,264
Another synaptic protein involved in the pathology of AD is contactin-2, a cell-adhesion protein that interacts with APP and beta-secretase 1 (BACE1). Chatterjee et al265 found that the protein was reduced in both brain tissue and CSF in AD. The less well-studied members of the synuclein family, beta-synuclein (β-syn) and gamma-synuclein (γ-syn), are also present in proteinaceous aggregates in some α-synucleinopathies.266 Oeckl et al167 was the first to measure all 3 synucleins protein family members, α, β, γ in CSF. They found increased concentrations of all synucleins in AD and CJD; however for PD, DLB, and atypical parkinsonian syndromes the concentrations were not altered. Furthermore, a high correlation between the 3 synucleins was seen. In another study by Oeckl et al,267 β-syn was quantified in blood and found it to be increased in AD and CJD compared with controls but not in other neurodegenerative diseases, such as PDD, DLB, amyotrophic lateral sclerosis (ALS), and FTD.
14-3-3
14-3-3 proteins refer to a family of 7 isoforms which are highly expressed in the brain, accounting for 1% of its soluble protein content. They are also particularly enriched at synapses (presynaptic) and important modulators of synaptic functions, such as neurotransmission and plasticity. 14-3-3 protein detection by Western blot has since long been used to detect CJD, albeit this technique is only semi-quantitative. However, more recently, 14-3-3 have been studied in the context of other neurodegenerative pathologies. 14-3-3 isoforms have not only been found to co-localize in LB in PD and NFTs in AD, but also been found to interact with key proteins such as tau and α-syn. They have also been genetically linked to both neurodegenerative diseases (PD, AD, and CJD) and neuropsychiatric disorders (schizophrenia and bipolar disorder).268,269 A recent study by Antonell et al270 found significantly increased gamma 14-3-3 concentrations in both FTD and AD compared with controls. For AD, increased concentrations were found already in a prodromal stage and the protein level was also significantly higher at later stages compared with FTD. Furthermore, when analyzing for 14-3-3, 96% of subjects were positive for neurodegeneration when applying the AT(N) system, compared with 94% for neurofilament light and 62% for neurogranin.270
Synaptophysin is one of the most used synaptic biomarkers in immunohistochemistry since it is the most abundant integral synaptic vesicle and plasma membrane protein. In studies of AD postmortem brain tissue, it has been shown that the synaptophysin content is reduced.271,272 Several studies have reported that the protein is not detectable in CSF,139,138,273 possibly due to its high hydrophobic profile.139 However, it has recently been reported to be detected in exosome preparations from body fluids.274,275
Neuronal-derived exosomes
A recent approach for the discovery of new synaptic biomarkers has been based on isolating neuronal exosomes from blood (plasma). As discussed previously, blood is an easily accessible peripheral fluid, preferred to CSF, which entails a more invasive extraction procedure. However, blood has the disadvantage of being further away from the brain and give peripheral contribution to the levels of the protein. Studying neuronal exosomes enriched from blood gives the advantage to use blood while hopefully better reflecting brain pathogenic processes. Explorative proteomic analysis has tried to map the protein content of the neuronal exosomes and confirmed the presence of several synaptic proteins such as Rab3a and GRIA4.276 In plasma samples, Goetzl et al274 reported significantly decreased neuronal-derived levels of synaptophysin together with synaptopodin, synaptotagmin-2, and neurogranin in patients with AD and FTD compared with controls. In the same study, GAP-43 and synapsin-1 were also detected, but were found to have significantly lower levels only in AD. Furthermore, in another study by Goetzl et al,277 plasma neuronal–derived exosome levels of NPTX2, neurexin 2, GRIA4, and neuroligin 1 were found to be significantly decreased in AD, where also GluR4 and neuroligin 1 correlated with cognitive loss. Another protein that has been quantified in neuronal exosomes is α-syn, found to have increased concentrations in PD compared with controls.278 For proteins such as neurogranin or α-syn, where peripheral expression complicates the quantification in blood, neuronally derived exosomes seem like an excellent option. However, even if this has promise, it is limited by expensive and time-consuming sample preparation, which as of today restricts its potential for high-throughput biomarker screening and its use in clinical routine. Nevertheless, exosomes are being connected to an increasing number of synaptopathies and they have even been implicated in the propagation of disease-associated proteins such as tau, Aβ, PrPC, and α-syn.279,280 They are a relatively unexplored source for synaptic biomarkers, which makes them a vital part of the field (Figure 2).
Figure 2.
Proteomic approaches in synaptic biomarkers discovery and validation. Proteomic studies can start with large explorative investigations in brain tissue, which might lead to the discovery of new candidate biomarkers. However, these studies can be seen as starting points, and they have no clinical utilities. Thus, investigations in CSF are needed to be able to translate the biomarker discovery into a tool of clinical use. Once the biomarker has been validated in CSF, further investigations can be carried in blood, a biofluid with higher accessibility and cheaper to use. On the other hand blood is further away from the brain and the targeted protein level might be susceptible to peripheral contribution, resulting in lower biomarker specificity and confounding results. A possible approach to overcome this problem is the use of plasma-derived neuronal exosomes. These investigations can be carried out with a targeted or non-targeted approach. In the diagram, pros and cons of both approaches are highlighted. ELISA indicates enzyme-linked immunosorbent assay; IP, immunoprecipitation; LC-MS, liquid chromatography-mass spectrometry; PRM, parallel reaction monitoring; SIMOA, single molecule array; WB, Western blot.
Figure made with www.biorender.com.
Conclusions and Future Perspective
Synapses are essential interconnecting points for neurons and are primarily affected in neurodegenerative and neurodevelopmental disorders.72 Accumulation of misfolded proteins seems to directly affect them,35 leading to their dysfunction and loss, which is closely related to the cognitive deficits seen in these aging disorders. This review summarizes latest studies on more established and newly investigated synaptic proteins as candidate biomarkers for synapse dysfunction and neuronal injury in different neurodegenerative diseases, in relation to both CSF and blood (Table 1).
Table 1.
Synaptic biomarkers changes in CSF and blood based on current literature.
Pathology Compared with controls | Cerebrospinal fluid | Blood | References | |||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
Amyloid-β | Prion | Lewy body | Tauopathies | Inflam. | Amyloid | Lewy body | ||||||
AD | CJD | PD | DLB | MSA | PSP | CBD | FTD | MS | AD | PD | ||
Presynaptic | ||||||||||||
SNAP-25 | ↑ | ↑ | ↑ | = | In CSF7,150-154,161 | |||||||
GAP-43 | ↑ | ↓ | = | ↓ | ↓E | In CSF109,143,144,145,147,161, in blood274 | ||||||
Synaptotagmin-1 | ↑ | = | In CSF159,161,158 | |||||||||
Alpha-synuclein | ||||||||||||
Total | ↓ | ↑ | ↓ | ↓ | ↓ | = | = | =S | ↑P, E↓R | In CSF163-169,171-178,180,207,281, in blood189,190,278 | ||
Oligomeric | = | ↑ | = | ↑S, P,R | In CSF169,174,197,198,207, in blood193,199-204 | |||||||
Phosporylated | = | ↑ | ↑ | ↑S | In CSF165,169,174,179,181,207, in blood185,203 | |||||||
Postsynaptic | ||||||||||||
Neurogranin | ↑ | ↑ | = | = | = | = | = | =P↓E | In CSF1,3,18,121,150,161,216,217,219,220,222,224-227 in blood121,220,274 | |||
Neuronal | ||||||||||||
Neurofilaments | ||||||||||||
Light chain | ↑ | ↑ | = | ↑ | ↑ | ↑ | ↑ | ↑ | ↑P, S | In CSF171,282-286, in blood132,134,135,287,288 | ||
Heavy chain | ↑ | In CSF286 | ||||||||||
Emerging | ||||||||||||
Synaptophysin | ↓E | In blood274 | ||||||||||
Synucleins | ||||||||||||
Gamma | ↑ | ↑ | = | = | In CSF167 | |||||||
Beta | ↑ | ↑ | = | = | ↑S | In CSF167, in blood167 | ||||||
Neuronal pentraxins | ||||||||||||
1 | ↓ | ↓ | ↓ | ↓ | In CSF231,246 | |||||||
2 | ↓ | ↓E | In CSF44,150,235,236,242, in blood277 | |||||||||
Receptor | ↓ | ↓ | ↓ | ↓ | ↓ | ↓ | In CSF237-239,244-246 | |||||
SV2A | ↓ | In CSF259 | ||||||||||
Contactin-2 | ↓ | In CSF265 | ||||||||||
Neurofascin | ↓ | In CSF231 | ||||||||||
Neurexin | ||||||||||||
1 | ↓ | In CSF231 | ||||||||||
2 | ↑ | ↓E | In CSF232, in blood277 | |||||||||
3 | ↑ | In CSF232 | ||||||||||
Syntaxin-1 | ↑ | In CSF232 | ||||||||||
Calsyntenin-1 | ↑ | In CSF232 | ||||||||||
Glutamate receptor 4 | ↑ | ↓E | In CSF232, in blood277 | |||||||||
Thy-1 membrane glycoprotein | ↑ | In CSF232 | ||||||||||
Synaptopodin | ↓E | In blood274 | ||||||||||
Synaptotagmin-2 | ↓E | In blood274 | ||||||||||
Synapsin 1 | ↓E | In blood274 | ||||||||||
Rab3a | = | In CSF153 | ||||||||||
Neuroligin 1 | ↓E | In blood277 | ||||||||||
14-3-3 Gamma | ↑ | ↑ | ↑ | In CSF270,289 | ||||||||
Granins | ↓ | In CSF231,264 |
Abbreviations: ↑, statistical increase; ↓, statistical decrease; =, no change; AD, Alzheimer’s disease; CBD, corticobasal degeneration; CJD, Creutzfeldt–Jakob disease; CSF, cerebrospinal fluid; DLB, dementia with Lewy body; E, plasma-derived exosomes; FTD, frontotemporal dementia; GAP-43, growth-associated protein 43; MS, multiple sclerosis; MSA, multiple system atrophy; P, plasma; PD, Parkinson’s disease; PSP, progressive supranuclear palsy; R, red blood cells (RBCs); S, serum; SNAP-25, synaptosomal-associated protein 25.
Current CSF synaptic biomarkers are altered in AD but seemingly not in other neurodegenerative disorders. This can reflect a higher response of synapses and neurons to Aβ-mediated damage, probably making AD the pathology with the highest synaptic damage. However, more efforts are needed to characterize synaptic loss in non-AD dementias and other synaptopathies. Increasing evidence suggests that synaptic dysfunction is also involved in neurodevelopmental diseases290,291 and neuropsychiatric disorders.26,292 Thus, the study of these conditions may help understanding differences or commonalities between synaptopathies.293
It can be noticed that most of the synaptic biomarkers described are represented by presynaptic proteins294 and, in AD, glutamatergic synapses appear to be primarily affected.6,294-297 Among the reviewed synaptic proteins, neurogranin is the most extensively studied and the evidence presented thus far is seemingly specific for AD or Aβ deposition. The other synaptic proteins also show changed levels in relation to AD, with most of them showing increased CSF concentrations, but also in non-AD neurodegenerative diseases (eg, PD, tauopathies), even though in these diseases they are less investigated. NfL is a good marker for general neuronal loss and it would be suitable to represent the “N” in the ATN criteria119,298 given that CSF t-tau also mainly changes in AD and CJD. Blood NfL strongly reflects CSF NfL.299 Elucidating the mechanisms of release of these proteins into biofluids would be of importance to understand their changes in concentration, thus connecting pathological mechanisms to biological responses and increase the interpretability of this biomarker category.
Understanding the pathological mechanisms responsible for synaptic damage is of central importance also during synaptic biomarker investigation. Brain studies could be a starting point, helping to understand the pathophysiological events and for selecting biomarker candidates. The next steps may involve the investigation of biofluids, like CSF, ideally followed by studies in blood, representing the way to bring the investigation further and possibly find synaptic biomarkers of clinical utility. The future of biomarkers ideally would be able to rely on sampling blood, which is a more accessible source than CSF. However, the possible contribution of peripheral expression of the biomarker protein, as discussed for neurogranin and α-syn, can represent a problem and, to date, we still have no blood biomarkers reflecting synaptic pathology. Neuronal-derived exosomes in blood can represent an alternative; however the complexity and variability of the exosome enrichment procedure is currently a drawback for large studies and routine use.
Future directions of research should consider more longitudinal studies, to compare protein time-related changes with the disease progression. The contribution of sex differences should be also considered in more detail, as developing evidence suggests that differing biomarker profiles do exist but is protein-specific.300,301
In conclusion, the available evidence on CSF synaptic biomarkers points toward the possible use of these proteins as indicators of synaptic alteration and elimination in synaptopathies, and their use to follow cognitive deficits in neurodegenerative diseases. More efforts are needed to assess their possible use in blood. Mechanistic studies will possibly help understanding how those proteins are affected in pathological processes thus increasing their value as potential biomarkers. Moreover, developing assays for their quantification using highly sensitive and high-throughput platforms will push synaptic protein quantification toward broader investigations. This overview of the field will hopefully highlight possible gaps and guide future studies.
Footnotes
Funding:The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: KB is supported by the Swedish Research Council (#2017-00915), the Alzheimer Drug Discovery Foundation (ADDF), USA (#RDAPB-201809-2016615), the Swedish Alzheimer Foundation (#AF-742881), Hjärnfonden, Sweden (#FO2017-0243), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986), and European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236). HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712), Swedish State Support for Clinical Research (#ALFGBG-720931), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), and the UK Dementia Research Institute at UCL.
Declaration of conflicting interests:The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: KB has served as a consultant, at advisory boards, or at data monitoring committees for Abcam, Axon, Biogen, Julius Clinical, Lilly, MagQu, Novartis, Roche Diagnostics, and Siemens Healthineers, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program. HZ has served at scientific advisory boards for Denali, Roche Diagnostics, Wave, Samumed, Siemens Healthineers, Pinteon Therapeutics, and CogRx; has given lectures in symposia sponsored by Fujirebio, Alzecure, and Biogen; and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program.
Author Contributions: EC, JN and NJA provided the initial idea and outline of content of the manuscript; EC created figures 1 and 2. JN created the table. All authors contributed to the content of the article and critically reviewed and edited the manuscript.
ORCID iD: Elena Camporesi
https://orcid.org/0000-0003-1044-0192
References
- 1. Taoufik E, Kouroupi G, Zygogianni O, Matsas R. Synaptic dysfunction in neurodegenerative and neurodevelopmental diseases: an overview of induced pluripotent stem-cell-based disease models. Open Biol. 2018;8:180138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Lepeta K, Lourenco MV, Schweitzer BC, et al. Synaptopathies: synaptic dysfunction in neurological disorders —a review from students to students. J Neurochem. 2016;138:785-805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Frankish H, Horton R. Prevention and management of dementia: a priority for public health. Lancet (London, England). 2017;390:2614-2615. [DOI] [PubMed] [Google Scholar]
- 4. Masliah E, Mallory M, Alford M, et al. Altered expression of synaptic proteins occurs early during progression of Alzheimer’s disease. Neurology. 2001;56:127-129. [DOI] [PubMed] [Google Scholar]
- 5. Janezic S, Threlfell S, Dodson PD, et al. Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc Natl Acad Sci U S A. 2013;110:E4016-E4025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Heurling K, Ashton NJ, Leuzy A, et al. Synaptic vesicle protein 2A as a potential biomarker in synaptopathies. Mol Cell Neurosci. 2019;97:34-42. [DOI] [PubMed] [Google Scholar]
- 7. Brinkmalm A, Brinkmalm G, Honer WG, et al. SNAP-25 is a promising novel cerebrospinal fluid biomarker for synapse degeneration in Alzheimer’s disease. Mol Neurodegener. 2014;9:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Korade Z, Mirnics K. Programmed to be human? Neuron. 2014;81:224-226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Ming GL, Song H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron. 2011;70:687-702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Sudhof TC. Towards an understanding of synapse formation. Neuron. 2018;100:276-293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Santos E, Noggle CA. Synaptic pruning. In: Goldstein S, Naglieri JA, eds. Encyclopedia of Child Behavior and Development. Boston, MA: Springer US; 2011:1464-1465. [Google Scholar]
- 12. Sando R, Bushong E, Zhu Y, et al. Assembly of excitatory synapses in the absence of glutamatergic neurotransmission. Neuron. 2017;94:312e313-312321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Südhof TC. Neurotransmitter release: the last millisecond in the life of a synaptic vesicle. Neuron. 2013;80:675-690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Hering H, Sheng M. Dendritic spines: structure, dynamics and regulation. Nat Rev Neurosci. 2001;2:880-888. [DOI] [PubMed] [Google Scholar]
- 15. Tao CL, Liu YT, Sun R, et al. Differentiation and characterization of excitatory and inhibitory synapses by cryo-electron tomography and correlative microscopy. J Neurosci. 2018;38:1493-1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Cooke SF, Bliss TV. Plasticity in the human central nervous system. Brain. 2006;129:1659-1673. [DOI] [PubMed] [Google Scholar]
- 17. Bosch M, Castro J, Saneyoshi T, Matsuno H, Sur M, Hayashi Y. Structural and molecular remodeling of dendritic spine substructures during long-term potentiation. Neuron. 2014;82:444-459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Piochon C, Kano M, Hansel C. LTD-like molecular pathways in developmental synaptic pruning. Nat Neurosci. 2016;19:1299-1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Vitureira N, Letellier M, Goda Y. Homeostatic synaptic plasticity: from single synapses to neural circuits. Curr Opin Neurobiol. 2012;22:516-521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Cardozo PL, de Lima IBQ, Maciel EMA, Silva NC, Dobransky T, Ribeiro FM. Synaptic elimination in neurological disorders. Curr Neuropharmacol. 2019;17:1071-1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Maiti P, Manna J, Ilavazhagan G, Rossignol J, Dunbar GL. Molecular regulation of dendritic spine dynamics and their potential impact on synaptic plasticity and neurological diseases. Neurosci Biobehav Rev. 2015;59:208-237. [DOI] [PubMed] [Google Scholar]
- 22. Kauer JA, Malenka RC. Synaptic plasticity and addiction. Nat Rev Neurosci. 2007;8:844-858. [DOI] [PubMed] [Google Scholar]
- 23. Guilherme N, Cooke SF, Bliss TVP. Synaptic plasticity, memory and the hippocampus. Nat Rev Neurosci. 2008;9:65-75. [DOI] [PubMed] [Google Scholar]
- 24. Boksa P. Abnormal synaptic pruning in schizophrenia: urban myth or reality? J Psychiatry Neurosci. 2012;37:75-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Racki V, Petric D, Kucic N, Grzeta N, Jurdana K, Roncevic-Grzeta I. Cortical gray matter loss in schizophrenia: could microglia be the culprit. Med Hypotheses. 2016;88:18-21. [DOI] [PubMed] [Google Scholar]
- 26. Osimo EF, Beck K, Reis Marques T, Howes OD. Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol Psychiatry. 2018;24:549-561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Bloom GS. Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 2014;71:505-508. [DOI] [PubMed] [Google Scholar]
- 28. Jack CR, Jr, Bennett DA, Blennow K, et al. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:535-562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Yoshiyama Y, Higuchi M, Zhang B, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53:337-351. [DOI] [PubMed] [Google Scholar]
- 30. Hamelin L, Lagarde J, Dorothee G, et al. Distinct dynamic profiles of microglial activation are associated with progression of Alzheimer’s disease. Brain. 2018;141:1855-1870. [DOI] [PubMed] [Google Scholar]
- 31. Dani M, Wood M, Mizoguchi R, et al. Microglial activation correlates in vivo with both tau and amyloid in Alzheimer’s disease. Brain. 2018;141:2740-2754. [DOI] [PubMed] [Google Scholar]
- 32. Liesz A. The vascular side of Alzheimer’s disease. Science. 2019;365:223-224. [DOI] [PubMed] [Google Scholar]
- 33. Jurcau A, Simion A. Oxidative stress in the pathogenesis of Alzheimer’s disease and cerebrovascular disease with therapeutic implications. CNS Neurol Disord Drug Targets. 2020;19:94-108. [DOI] [PubMed] [Google Scholar]
- 34. Terry RD, Masliiah E, Salmon DP, et al. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572-580. [DOI] [PubMed] [Google Scholar]
- 35. Yu W, Lu B. Synapses and dendritic spines as pathogenic targets in Alzheimer’s disease. Neural Plast. 2012;2012:247150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Blennow K, Bogdanovic N, Alafuzoff I, Ekman R, Davidsson P. Synaptic pathology in Alzheimer’s disease: relation to severity of dementia, but not to senile plaques, neurofibrillary tangles, or the ApoE4 allele. J Neural Transm (Vienna). 1996;103:603-618. [DOI] [PubMed] [Google Scholar]
- 37. Selkoe DJ. Alzheimer’s disease is a synaptic failure. Science. 2002;298:789-791. [DOI] [PubMed] [Google Scholar]
- 38. Masliah E, Mallory M, Alford M, et al. Altered expression of synaptic proteins occurs early during progression of Alzheimer’s disease. Neurology. 2001;56:127-129. [DOI] [PubMed] [Google Scholar]
- 39. Bertoni-Freddari C, Fattoretti P, Casoli T, Meier-Ruge W, Ulrich J. Morphological adaptive response of the synaptic junctional zones in the human dentate gyrus during aging and Alzheimer’s disease. Brain Res. 1990;517:69-75. [DOI] [PubMed] [Google Scholar]
- 40. Scheff SW, Price DA. Synapse loss in the temporal lobe in Alzheimer’s disease. Ann Neurol. 1993;33:190-199. [DOI] [PubMed] [Google Scholar]
- 41. Reddy PH, Mani G, Park BS, et al. Differential loss of synaptic proteins in Alzheimer’s disease: implications for synaptic dysfunction. J Alzheimers Dis. 2005;7:103-117; discussion 173. [DOI] [PubMed] [Google Scholar]
- 42. Clare R, King VG, Wirenfeldt M, Vinters HV. Synapse loss in dementias. J Neurosci Res. 2010;88:2083-2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Scheff SW, Price DA, Schmitt FA, Mufson EJ. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging. 2006;27:1372-1384. [DOI] [PubMed] [Google Scholar]
- 44. Scheff SW, Price DA, Schmitt FA, DeKosky ST, Mufson EJ. Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology. 2007;68:1501-1508. [DOI] [PubMed] [Google Scholar]
- 45. Wang Z, Jackson RJ, Hong W, et al. Human brain-derived Aβ oligomers bind to synapses and disrupt synaptic activity in a manner that requires APP. J Neurosci. 2017;37:11947-11966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Walsh DM, Klyubin I, Fadeeva JV, et al. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535-539. [DOI] [PubMed] [Google Scholar]
- 47. Tu S, Okamoto S, Lipton SA, Xu H. Oligomeric Abeta-induced synaptic dysfunction in Alzheimer’s disease. Mol Neurodegener. 2014;9:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Guerrero-Muñoz MJ, Gerson J, Castillo-Carranza DL. Tau oligomers: the toxic player at synapses in Alzheimer’s disease. Front Cell Neurosci. 2015;9:464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Colom-Cadena M, Spires-Jones T, Zetterberg H, et al. The clinical promise of biomarkers of synapse damage or loss in Alzheimer’s disease. Alzheimers Res Ther. 2020;12:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Wang HW, Pasternak JF, Kuo H, et al. Soluble oligomers of beta amyloid (1-42) inhibit long-term potentiation but not long-term depression in rat dentate gyrus. Brain Res. 2002;924:133-140. [DOI] [PubMed] [Google Scholar]
- 51. Lambert MP, Barlow AK, Chromy BA, et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998;95:6448-6453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, Sabatini BL. Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J Neurosci. 2007;27:2866-2875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Cline EN, Bicca MA, Viola KL, Klein WL. The amyloid-beta oligomer hypothesis: beginning of the third decade. J Alzheimers Dis. 2018;64:S567-S610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Fein JA, Sokolow S, Miller CA, et al. Co-localization of amyloid beta and tau pathology in Alzheimer’s disease synaptosomes. Am J Pathol. 2008;172:1683-1692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Takahashi RH, Capetillo-Zarate E, Lin MT, Milner TA, Gouras GK. Co-occurrence of Alzheimer’s disease ss-amyloid and tau pathologies at synapses. Neurobiol Aging. 2010;31:1145-1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Smith LM, Strittmatter SM. Binding sites for amyloid-β oligomers and synaptic toxicity. Cold Spring Harb Perspect Med. 2017;7:a024075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Younan ND, Sarell CJ, Davies P, Brown DR, Viles JH. The cellular prion protein traps Alzheimer’s Abeta in an oligomeric form and disassembles amyloid fibers. FASEB J. 2013;27:1847-1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Brito-Moreira J, Lourenco MV, Oliveira MM, et al. Interaction of amyloid-beta (Abeta) oligomers with neurexin 2alpha and neuroligin 1 mediates synapse damage and memory loss in mice. J Biol Chem. 2017;292:7327-7337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Cissé M, Halabisky B, Harris J, et al. Reversing EphB2 depletion rescues cognitive functions in Alzheimer model. Nature. 2011;469:47-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Ding Y, Zhao J, Zhang X, et al. Amyloid beta oligomers target to extracellular and intracellular neuronal synaptic proteins in Alzheimer’s disease. Front Neurol. 2019;10:1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Wei W, Nguyen LN, Kessels HW, Hagiwara H, Sisodia S, Malinow R. Amyloid beta from axons and dendrites reduces local spine number and plasticity. Nat Neurosci. 2010;13:190-196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Puzzo D, Piacentini R, Fa M, et al. LTP and memory impairment caused by extracellular Abeta and Tau oligomers is APP-dependent. eLife. 2017;6:e26991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Jadhav S, Cubinkova V, Zimova I, et al. Tau-mediated synaptic damage in Alzheimer’s disease. Transl Neurosci. 2015;6:214-226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Guo JL, Lee VM. Cell-to-cell transmission of pathogenic proteins in neurodegenerative diseases. Nat Med. 2014;20:130-138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Nath S, Agholme L, Kurudenkandy FR, Granseth B, Marcusson J, Hallbeck M. Spreading of neurodegenerative pathology via neuron-to-neuron transmission of beta-amyloid. J Neurosci. 2012;32:8767-8777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Braak H, Del Tredici K. Alzheimer’s pathogenesis: is there neuron-to-neuron propagation. Acta Neuropathol. 2011;121:589-595. [DOI] [PubMed] [Google Scholar]
- 67. Walsh DM, Selkoe DJ. A critical appraisal of the pathogenic protein spread hypothesis of neurodegeneration. Nat Rev Neurosci. 2016;17:251-260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. de Calignon A, Polydoro M, Suárez-Calvet M, et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron. 2012;73:685-697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Clavaguera F, Akatsu H, Fraser G, et al. Brain homogenates from human tauopathies induce tau inclusions in mouse brain. Proc Natl Acad Sci U S A. 2013;110:9535-9540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Orr ME, Sullivan AC, Frost B. A brief overview of tauopathy: causes, consequences, and therapeutic strategies. Trends Pharmacol Sci. 2017;38:637-648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Kovacs GG. Molecular pathological classification of neurodegenerative diseases: turning towards precision medicine. Int J Mol Sci. 2016;17:189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Vogels T, Murgoci AN, Hromadka T. Intersection of pathological tau and microglia at the synapse. Acta Neuropathol Commun. 2019;7:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Dejanovic B, Huntley MA, De Maziere A, et al. Changes in the synaptic proteome in tauopathy and rescue of tau-induced synapse loss by C1q antibodies. Neuron. 2018;100:1322-1336.e7. [DOI] [PubMed] [Google Scholar]
- 74. Jackson JS, Witton J, Johnson JD, et al. Altered synapse stability in the early stages of tauopathy. Cell Rep. 2017;18:3063-3068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Goedert M, Jakes R, Spillantini MG. The synucleinopathies: twenty years on. J Parkinsons Dis. 2017;7:S51-S69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Baba M, Nakajo S, Tu PH, et al. Aggregation of alpha-synuclein in Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy bodies. Am J Pathol. 1998;152:879-884. [PMC free article] [PubMed] [Google Scholar]
- 77. Brás IC, Dominguez-Meijide A, Gerhardt E, et al. Synucleinopathies: where we are and where we need to go. J Neurochem. 2020;153:433-454. [DOI] [PubMed] [Google Scholar]
- 78. Sanderson JB, De S, Jiang H, et al. Analysis of alpha-synuclein species enriched from cerebral cortex of humans with sporadic dementia with Lewy bodies. Brain Commun. 2020;2:fcaa010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Aarsland D, Beyer MK, Kurz MW. Dementia in Parkinson’s disease. Curr Opin Neurol. 2008;21:676-682. [DOI] [PubMed] [Google Scholar]
- 80. Murphy DD, Rueter SM, Trojanowski JQ, Lee VM. Synucleins are developmentally expressed, and a-synuclein regulates the size of the presynaptic vesicular pool in primary hippocampal neurons. J Neurosci. 2000;20:3214-3220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Chandra S, Chen X, Rizo J, Jahn R, Sudhof TC. A broken alpha -helix in folded alpha -Synuclein. J Biol Chem. 2003;278:15313-15318. [DOI] [PubMed] [Google Scholar]
- 82. Hawk BJD, Khounlo R, Shin YK. Alpha-synuclein continues to enhance SNARE-dependent vesicle docking at exorbitant concentrations. Front Neurosci. 2019;13:216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Burre J, Sharma M, Tsetsenis T, Buchman V, Etherton MR, Sudhof TC. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science. 2010;329:1663-1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Pozo Devoto VM, Falzone TL. Mitochondrial dynamics in Parkinson’s disease: a role for alpha-synuclein? Dis Model Mech. 2017;10:1075-1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Cooper AA, Gitler AD, Cashikar A, et al. Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson’s models. Science. 2006;313:324-328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Wang X, Becker K, Levine N, et al. Pathogenic alpha-synuclein aggregates preferentially bind to mitochondria and affect cellular respiration. Acta Neuropathol Commun. 2019;7:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Bernal-Conde LD, Ramos-Acevedo R, Reyes-Hernandez MA, et al. Alpha-synuclein physiology and pathology: a perspective on cellular structures and organelles. Front Neurosci. 2020;13:1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Loov C, Scherzer CR, Hyman BT, Breakefield XO, Ingelsson M. alpha-synuclein in extracellular vesicles: functional implications and diagnostic opportunities. Cell Mol Neurobiol. 2016;36:437-448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Forloni G, Artuso V, La Vitola P, Balducci C. Oligomeropathies and pathogenesis of Alzheimer and Parkinson’s diseases. Mov Disord. 2016;31:771-781. [DOI] [PubMed] [Google Scholar]
- 90. Prots I, Grosch J, Brazdis RM, et al. alpha-Synuclein oligomers induce early axonal dysfunction in human iPSC-based models of synucleinopathies. Proc Natl Acad Sci U S A. 2018;115:7813-7818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Tarutani A, Arai T, Murayama S, Hisanaga SI, Hasegawa M. Potent prion-like behaviors of pathogenic alpha-synuclein and evaluation of inactivation methods. Acta Neuropathol Commun. 2018;6:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Ma J, Gao J, Wang J, Xie A. Prion-like mechanisms in Parkinson’s disease. Front Neurosci. 2019;13:552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Jellinger KA, Lantos PL. Papp-Lantos inclusions and the pathogenesis of multiple system atrophy: an update. Acta Neuropathol. 2010;119:657-667. [DOI] [PubMed] [Google Scholar]
- 94. Peng C, Gathagan RJ, Covell DJ, et al. Cellular milieu imparts distinct pathological alpha-synuclein strains in alpha-synucleinopathies. Nature. 2018;557:558-563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Savica R, Grossardt BR, Bower JH, et al. Survival and causes of death among people with clinically diagnosed synucleinopathies with parkinsonism: a population-based study. JAMA Neurol. 2017;74:839-846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Sekiya H, Kowa H, Koga H, et al. Wide distribution of alpha-synuclein oligomers in multiple system atrophy brain detected by proximity ligation. Acta Neuropathol. 2019;137:455-466. [DOI] [PubMed] [Google Scholar]
- 97. Schweighauser M, Shi Y, Tarutani A, et al. Structures of α-synuclein filaments from multiple system atrophy [published online ahead of print May 27, 2020]. Nature. doi: 10.1038/s41586-020-2317-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Zaccai J, McCracken C, Brayne C. A systematic review of prevalence and incidence studies of dementia with Lewy bodies. Age Ageing. 2005;34:561-566. [DOI] [PubMed] [Google Scholar]
- 99. Bellucci A, Mercuri NB, Venneri A, et al. Review: Parkinson’s disease: from synaptic loss to connectome dysfunction. Neuropathol Appl Neurobiol. 2016;42:77-94. [DOI] [PubMed] [Google Scholar]
- 100. Uéda K, Fukushima H, Masliah E, et al. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc Natl Acad Sci U S A. 1993;90:11282-11286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Hamilton RL. Lewy bodies in Alzheimer’s disease: a neuropathological review of 145 cases using alpha-synuclein immunohistochemistry. Brain Pathol. 2000;10:378-384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Kovacs GG, Milenkovic I, Wöhrer A, et al. Non-Alzheimer neurodegenerative pathologies and their combinations are more frequent than commonly believed in the elderly brain: a community-based autopsy series. Acta Neuropathol. 2013;126:365-384. [DOI] [PubMed] [Google Scholar]
- 103. Beach TG, Monsell SE, Phillips LE, Kukull W. Accuracy of the clinical diagnosis of Alzheimer disease at National Institute on Aging Alzheimer Disease Centers, 2005-2010. J Neuropathol Exp Neurol. 2012;71:266-273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Moussaud S, Jones DR, Moussaud-Lamodière EL, Delenclos M, Ross OA, McLean PJ. Alpha-synuclein and tau: teammates in neurodegeneration? Mol Neurodegener. 2014;9:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Wenning GK, Krismer F, Poewe W. New insights into atypical parkinsonism. Curr Opin Neurol. 2011;24:331-338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Olsson B, Lautner R, Andreasson U, et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 2016;15:673-684. [DOI] [PubMed] [Google Scholar]
- 107. Dubois B, Feldman HH, Jacova C, et al. Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. 2014;13:614-629. [DOI] [PubMed] [Google Scholar]
- 108. Oeckl P, Steinacker P, Feneberg E, Otto M. Neurochemical biomarkers in the diagnosis of frontotemporal lobar degeneration: an update. J Neurochem. 2016;138:184-192. [DOI] [PubMed] [Google Scholar]
- 109. Sjogren M, Minthon L, Davidsson P, et al. CSF levels of tau, beta-amyloid(1-42) and GAP-43 in frontotemporal dementia, other types of dementia and normal aging. J Neural Transm (Vienna). 2000;107:563-579. [DOI] [PubMed] [Google Scholar]
- 110. Riemenschneider M, Wagenpfeil S, Vanderstichele H, et al. Phospho-tau/total tau ratio in cerebrospinal fluid discriminates Creutzfeldt-Jakob disease from other dementias. Mol Psychiatry. 2003;8:343-347. [DOI] [PubMed] [Google Scholar]
- 111. Otto M, Wiltfang J, Tumania H, et al. Elevated levels of tau-protein in cerebrospinal fluid of patients with Creutzfeldt–Jakob disease. Neurosci Lett. 1997;225:210-212. [DOI] [PubMed] [Google Scholar]
- 112. Frisoni GB, Fox NC, Jack CR, Jr, Scheltens P, Thompson PM. The clinical use of structural MRI in Alzheimer disease. Nat Rev Neurol. 2010;6:67-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Barthel H, Sabri O. Clinical use and utility of amyloid imaging. J Nucl Med. 2017;58:1711-1717. [DOI] [PubMed] [Google Scholar]
- 114. Filippi L, Chiaravalloti A, Bagni O, Schillaci O. (18)F-labeled radiopharmaceuticals for the molecular neuroimaging of amyloid plaques in Alzheimer’s disease. Am J Nucl Med Mol Imaging. 2018;8:268-281. [PMC free article] [PubMed] [Google Scholar]
- 115. Schöll M, Maass A, Mattsson N, et al. Biomarkers for tau pathology. Mol Cell Neurosci. 2019;97:18-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Choo IH, Ni R, Schöll M, Wall A, Almkvist O, Nordberg A. Combination of 18F-FDG PET and cerebrospinal fluid biomarkers as a better predictor of the progression to Alzheimer’s disease in mild cognitive impairment patients. J Alzheimers Dis. 2013;33:929-939. [DOI] [PubMed] [Google Scholar]
- 117. Schöll M, Damián A, Engler H. Fluorodeoxyglucose PET in neurology and psychiatry. PET Clin. 2014;9:371-390. [DOI] [PubMed] [Google Scholar]
- 118. Mecca AP, Chen MK, O’Dell RS, et al. In vivo measurement of widespread synaptic loss in Alzheimer’s disease with SV2A PET. Alzheimers Dement. 2020;16:974-982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Jack CR, Bennett DA, Blennow K, et al. A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology. 2016;87:539-547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Ashton NJ, Nevado-Holgado AJ, Barber IS, et al. A plasma protein classifier for predicting amyloid burden for preclinical Alzheimer’s disease. Sci Adv. 2019;5:eaau7220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Kvartsberg H, Portelius E, Andreasson U, et al. Characterization of the postsynaptic protein neurogranin in paired cerebrospinal fluid and plasma samples from Alzheimer’s disease patients and healthy controls. Alzheimers Res Ther. 2015;7:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Nakamura A, Kaneko N, Villemagne VL, et al. High performance plasma amyloid-beta biomarkers for Alzheimer’s disease. Nature. 2018;554:249-254. [DOI] [PubMed] [Google Scholar]
- 123. Schindler SE, Bollinger JG, Ovod V, et al. High-precision plasma beta-amyloid 42/40 predicts current and future brain amyloidosis. Neurology. 2019;93:e1647-e1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Janelidze S, Stomrud E, Palmqvist S, et al. Plasma β-amyloid in Alzheimer’s disease and vascular disease. Sci Rep. 2016;6:1-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Palmqvist S, Janelidze S, Stomrud E, et al. Performance of fully automated plasma assays as screening tests for Alzheimer disease–related β-amyloid status. JAMA Neurol. 2019;76:1060-1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Mielke MM, Hagen CE, Xu J, et al. Plasma phospho-tau181 increases with Alzheimer’s disease clinical severity and is associated with tau-and amyloid-positron emission tomography. Alzheimers Dement. 2018;14:989-997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Janelidze S, Mattsson N, Palmqvist S, et al. Plasma P-tau181 in Alzheimer’s disease: relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s dementia. Nat Med. 2020;26:379-386. [DOI] [PubMed] [Google Scholar]
- 128. Thijssen EH, La Joie R, Wolf A, et al. Diagnostic value of plasma phosphorylated tau181 in Alzheimer’s disease and frontotemporal lobar degeneration. Nat Med. 2020;26:387-397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Karikari TK, Pascoal TA, Ashton NJ, et al. Blood phosphorylated tau 181 as a biomarker for Alzheimer’s disease: a diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol. 2020;19:422-433. [DOI] [PubMed] [Google Scholar]
- 130. Mattsson N, Zetterberg H, Janelidze S, et al. Plasma tau in Alzheimer disease. Neurology. 2016;87:1827-1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Khalil M, Teunissen CE, Otto M, et al. Neurofilaments as biomarkers in neurological disorders. Nat Rev Neurol. 2018;14:577-589. [DOI] [PubMed] [Google Scholar]
- 132. Hansson O, Janelidze S, Hall S, et al. Blood-based NfL: a biomarker for differential diagnosis of parkinsonian disorder. Neurology. 2017;88:930-937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Mattsson N, Cullen NC, Andreasson U, Zetterberg H, Blennow K. Association between longitudinal plasma neurofilament light and neurodegeneration in patients with Alzheimer disease. JAMA Neurol. 2019;76:791-799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Ashton NJ, Leuzy A, Lim YM, et al. Increased plasma neurofilament light chain concentration correlates with severity of post-mortem neurofibrillary tangle pathology and neurodegeneration. Acta Neuropathol Commun. 2019;7:1-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Mattsson N, Andreasson U, Zetterberg H, Blennow K. Association of plasma neurofilament light with neurodegeneration in patients with Alzheimer disease. JAMA Neurol. 2017;74:557-566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Zetterberg H, Skillbäck T, Mattsson N, et al. Association of cerebrospinal fluid neurofilament light concentration with Alzheimer disease progression. JAMA Neurol. 2016;73:60-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Benedet Al, Ashton NJ, Pascoal TA, et al. Plasma neurofilament light associates with Alzheimer’s disease metabolic decline in amyloid-positive individuals. Alzheimers Dement (Amst). 2019;11:679-689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Davidsson P, Jahn R, Bergquist J, Ekman R, Blennow K. Synaptotagmin, a synaptic vesicle protein, is present in human cerebrospinal fluid. Mol Chem Neuropathol. 1996;27:195-210. [DOI] [PubMed] [Google Scholar]
- 139. Davidsson P, Puchades M, Blennow K. Identification of synaptic vesicle, pre- and postsynaptic proteins in human cerebrospinal fluid using liquid-phase isoelectric focusing. Electrophoresis. 1999;20:431-437. [DOI] [PubMed] [Google Scholar]
- 140. Benowitz LI, Routtenberg A. GAP-43: an intrinsic determinant of neuronal development and plasticity. Trends Neurosci. 1997;20:84-91. [DOI] [PubMed] [Google Scholar]
- 141. Holahan MR. A shift from a pivotal to supporting role for the growth-associated protein (GAP-43) in the coordination of axonal structural and functional plasticity. Front Cell Neurosci. 2017;11:266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Sjögren M, Davidsson P, Gottfries J, et al. The cerebrospinal fluid levels of tau, growth-associated protein-43 and soluble amyloid precursor protein correlate in Alzheimer’s disease, reflecting a common pathophysiological process. Dement Geriatr Cogn Disord. 2001;12:257-264. [DOI] [PubMed] [Google Scholar]
- 143. Sandelius Å, Portelius E, Källén Å, et al. Elevated CSF GAP-43 is Alzheimer’s disease specific and associated with tau and amyloid pathology. Alzheimers Dement. 2019;15:55-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Remnestal J, Just D, Mitsios N, et al. CSF profiling of the human brain enriched proteome reveals associations of neuromodulin and neurogranin to Alzheimer’s disease. Proteomics Clin Appl. 2016;10:1242-1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Haggmark A, Bystrom S, Ayoglu B, et al. Antibody-based profiling of cerebrospinal fluid within multiple sclerosis. Proteomics. 2013;13:2256-2267. [DOI] [PubMed] [Google Scholar]
- 146. Rot U, Sandelius A, Emersic A, Zetterberg H, Blennow K. Cerebrospinal fluid GAP-43 in early multiple sclerosis. Mult Scler J Exp Transl Clin. 2018;4:2055217318792931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Sandelius A, Cullen NC, Kallen A, et al. Transient increase in CSF GAP-43 concentration after ischemic stroke. BMC Neurol. 2018;18:202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Südhof TC. The synaptic vesicle cycle. Annu Rev Neurosci. 2004;27:509-547. [DOI] [PubMed] [Google Scholar]
- 149. Antonucci F, Corradini I, Fossati G, Tomasoni R, Menna E, Matteoli M. SNAP-25, a known presynaptic protein with emerging postsynaptic functions. Front Synaptic Neurosci. 2016;8:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Galasko D, Xiao M, Xu D, et al. Synaptic biomarkers in CSF aid in diagnosis, correlate with cognition and predict progression in MCI and Alzheimer’s disease. Alzheimers Dement (N Y). 2019;5:871-882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Wang S, Zhang J, Pan T; for Alzheimer’s Disease Neuroimaging Initiative. APOE epsilon4 is associated with higher levels of CSF SNAP-25 in prodromal Alzheimer’s disease. Neurosci Lett. 2018;685:109-113. [DOI] [PubMed] [Google Scholar]
- 152. Zhang H, Therriault J, Kang MS, et al. Cerebrospinal fluid synaptosomal-associated protein 25 is a key player in synaptic degeneration in mild cognitive impairment and Alzheimer’s disease. Alzheimers Res Ther. 2018;10:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Bereczki E, Bogstedt A, Höglund K, et al. Synaptic proteins in CSF relate to Parkinson’s disease stage markers. NPJ Parkinsons Dis. 2017;3:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Wang C, Zhao D, Shah SZA, Yang W, Li C, Yang L. Proteome analysis of potential synaptic vesicle cycle biomarkers in the cerebrospinal fluid of patients with sporadic Creutzfeldt–Jakob disease. Mol Neurobiol. 2017;54:5177-5191. [DOI] [PubMed] [Google Scholar]
- 155. Barakauskas VE, Moradian A, Barr AM, et al. Quantitative mass spectrometry reveals changes in SNAP-25 isoforms in schizophrenia. Schizophr Res. 2016;177:44-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Courtney NA, Bao H, Briguglio JS, Chapman ER. Synaptotagmin 1 clamps synaptic vesicle fusion in mammalian neurons independent of complexin. Nat Commun. 2019;10:4076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Park Y, Ryu JK. Models of synaptotagmin-1 to trigger Ca(2+) -dependent vesicle fusion. FEBS Lett. 2018;592:3480-3492. [DOI] [PubMed] [Google Scholar]
- 158. Öhrfelt A, Brinkmalm A, Dumurgier J, et al. The pre-synaptic vesicle protein synaptotagmin is a novel biomarker for Alzheimer’s disease. Alzheimers Res Ther. 2016;8:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Tible M, Sandelius Höglund ÅK, et al. Dissection of synaptic pathways through the analysis of cerebrospinal fluid biomarkers: a combined tool for predicting Alzheimer’s disease [published online ahead of print June 25, 2020]. Neurology. doi: 10.1212/WNL.0000000000010131. [DOI] [Google Scholar]
- 160. Ohrfelt A, Brinkmalm A, Dumurgier J, et al. A novel ELISA for the measurement of cerebrospinal fluid SNAP-25 in patients with Alzheimer’s disease. Neuroscience. 2019;420:136-144. [DOI] [PubMed] [Google Scholar]
- 161. Clarke MTM, Brinkmalm A, Foiani MS, et al. CSF synaptic protein concentrations are raised in those with atypical Alzheimer’s disease but not frontotemporal dementia. Alzheimers Res Ther. 2019;11:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Lavedan C. The synuclein family. Genome Res. 2020;8:871-880. [DOI] [PubMed] [Google Scholar]
- 163. Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel-Döring F, Trenkwalder C, Schlossmacher MG. α-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: a cohort study. Lancet Neurol. 2011;10:230-240. [DOI] [PubMed] [Google Scholar]
- 164. Tokuda T, Salem SA, Allsop D, et al. Decreased α-synuclein in cerebrospinal fluid of aged individuals and subjects with Parkinson’s disease. Biochem Bioph Res Co. 2006;349:162-166. [DOI] [PubMed] [Google Scholar]
- 165. Mondello S, Constantinescu R, Zetterberg H, Andreasson U, Holmberg B, Jeromin A. CSF α-synuclein and UCH-L1 levels in Parkinson’s disease and atypical parkinsonian disorders. Parkinsonism Relat Disord. 2014;20:382-387. [DOI] [PubMed] [Google Scholar]
- 166. Öhrfelt A, Grognet P, Andreasen N, et al. Cerebrospinal fluid α-synuclein in neurodegenerative disorders—a marker of synapse loss? Neurosci Lett. 2009;450:332-335. [DOI] [PubMed] [Google Scholar]
- 167. Oeckl P, Metzger F, Nagl M, et al. Alpha-, Beta-, and Gamma-synuclein quantification in cerebrospinal fluid by multiple reaction monitoring reveals increased concentrations in Alzheimer’s and Creutzfeldt-Jakob disease but no alteration in synucleinopathies. Mol Cell Proteomics. 2016;15:3126-3138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Gao L, Tang H, Nie K, et al. Cerebrospinal fluid alpha-synuclein as a biomarker for Parkinson’s disease diagnosis: a systematic review and meta-analysis. Int J Neurosci. 2015;125:645-654. [DOI] [PubMed] [Google Scholar]
- 169. Eusebi P, Giannandrea D, Biscetti L, et al. Diagnostic utility of cerebrospinal fluid α-synuclein in Parkinson’s disease: a systematic review and meta-analysis. Mov Disord. 2017;32:1389-1400. [DOI] [PubMed] [Google Scholar]
- 170. Zhou B, Wen M, Yu W-F, Zhang C-L, Jiao L. The diagnostic and differential diagnosis utility of cerebrospinal fluid α-synuclein levels in Parkinson’s disease: a meta-analysis. Parkinsons Dis. 2015;2015;567386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Hall S, Öhrfelt A, Constantinescu R, et al. Accuracy of a panel of 5 cerebrospinal fluid biomarkers in the differential diagnosis of patients with dementia and/or parkinsonian disorders. Arch Neurol. 2012;69:1445-1452. [DOI] [PubMed] [Google Scholar]
- 172. Tateno F, Sakakibara R, Kawai T, Kishi M, Murano T. Alpha-synuclein in the cerebrospinal fluid differentiates synucleinopathies (Parkinson disease, dementia with Lewy bodies, multiple system atrophy) from Alzheimer disease. Alzheimer Dis Assoc Disord. 2012;26:213-216. [DOI] [PubMed] [Google Scholar]
- 173. Wennström M, Surova Y, Hall S, et al. Low CSF levels of both α-synuclein and the α-synuclein cleaving enzyme neurosin in patients with synucleinopathy. PLoS ONE. 2013;8:e53250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Majbour NK, Chiasserini D, Vaikath NN, et al. Increased levels of CSF total but not oligomeric or phosphorylated forms of alpha-synuclein in patients diagnosed with probable Alzheimer’s disease. Sci Rep. 2017;7:1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Wang Z-Y, Han Z-M, Liu Q-F, Tang W, Ye K, Yao Y-Y. Use of CSF α-synuclein in the differential diagnosis between Alzheimer’s disease and other neurodegenerative disorders. Int Psychogeriatr. 2015;27:1429-1438. [DOI] [PubMed] [Google Scholar]
- 176. Berge G, Sando SB, Albrektsen G, et al. Alpha-synuclein measured in cerebrospinal fluid from patients with Alzheimer’s disease, mild cognitive impairment, or healthy controls: a two year follow-up study. BMC Neurol. 2016;16:180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Kapaki E, Paraskevas GP, Emmanouilidou E, Vekrellis K. The diagnostic value of CSF α-synuclein in the differential diagnosis of dementia with Lewy bodies vs. PLoS ONE. 2013;8:e81654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Llorens F, Kruse N, Schmitz M, et al. Evaluation of α-synuclein as a novel cerebrospinal fluid biomarker in different forms of prion diseases. Alzheimers Dement. 2017;13:710-719. [DOI] [PubMed] [Google Scholar]
- 179. Wang H, Stewart T, Toledo JB, et al. A longitudinal study of Total and phosphorylated α-Synuclein with other biomarkers in cerebrospinal fluid of Alzheimer’s disease and mild cognitive impairment. J Alzheimers Dis. 2018;61:1541-1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Llorens F, Kruse N, Karch A, et al. Validation of α-synuclein as a CSF biomarker for sporadic Creutzfeldt-Jakob disease. Mol Neurobiol. 2018;55:2249-2257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Schmitz M, Villar-Piqué A, Llorens F, et al. Cerebrospinal fluid total and phosphorylated α-synuclein in patients with Creutzfeldt–Jakob disease and synucleinopathy. Mol Neurobiol. 2019;56:3476-3483. [DOI] [PubMed] [Google Scholar]
- 182. Mollenhauer B, Bowman FD, Drake D, et al. Antibody-based methods for the measurement of α-synuclein concentration in human cerebrospinal fluid–method comparison and round robin study. J Neurochem. 2019;149:126-138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Barbour R, Kling K, Anderson JP, et al. Red blood cells are the major source of alpha-synuclein in blood. Neurodegener Dis. 2008;5:55-59. [DOI] [PubMed] [Google Scholar]
- 184. Barkovits K, Kruse N, Linden A, et al. Blood contamination in CSF and its impact on quantitative analysis of alpha-synuclein. Cells. 2020;9:370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Foulds PG, Diggle P, Mitchell JD, et al. A longitudinal study on α-synuclein in blood plasma as a biomarker for Parkinson’s disease. Sci Rep. 2013;3:2540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Ishii R, Tokuda T, Tatebe H, et al. Decrease in plasma levels of α-synuclein is evident in patients with Parkinson’s disease after elimination of heterophilic antibody interference. PLoS ONE. 2015;10:e0123162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Malec-Litwinowicz M, Plewka A, Plewka D, et al. The relation between plasma α-synuclein level and clinical symptoms or signs of Parkinson’s disease. Neurol I Neurochir Pol. 2018;52:243-251. [DOI] [PubMed] [Google Scholar]
- 188. Fan Z, Pan Y-T, Zhang Z-Y, et al. Systemic activation of NLRP3 inflammasome and plasma α-synuclein levels are correlated with motor severity and progression in Parkinson’s disease. J Neuroinflammation. 2020;17:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Bougea A, Stefanis L, Paraskevas GP, Emmanouilidou E, Vekrelis K, Kapaki E. Plasma alpha-synuclein levels in patients with Parkinson’s disease: a systematic review and meta-analysis. Neurol Sci. 2019;40:929-938. [DOI] [PubMed] [Google Scholar]
- 190. Laske C, Fallgatter AJ, Stransky E, Hagen K, Berg D, Maetzler W. Decreased α-synuclein serum levels in patients with Lewy body dementia compared to Alzheimer’s disease patients and control subjects. Dement Geriatr Cogn Disord. 2011;31:413-416. [DOI] [PubMed] [Google Scholar]
- 191. Abd-Elhadi S, Honig A, Simhi-Haham D, et al. Total and proteinase K-resistant α-synuclein levels in erythrocytes, determined by their ability to bind phospholipids, associate with Parkinson’s disease. Sci Rep. 2015;5:11120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Baldacci F, Daniele S, Piccarducci R, et al. Potential diagnostic value of red blood cells α-synuclein heteroaggregates in Alzheimer’s disease. Mol Neurobiol. 2019;56:6451-6459. [DOI] [PubMed] [Google Scholar]
- 193. Daniele S, Frosini D, Pietrobono D, et al. α-synuclein heterocomplexes with β-amyloid are increased in red blood cells of Parkinson’s disease patients and correlate with disease severity. Front Mol Neurosci. 2018;11:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Kang W, Chen W, Yang Q, et al. Salivary total α-synuclein, oligomeric α-synuclein and SNCA variants in Parkinson’s disease patients. Sci Rep. 2016;6:28143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Devic I, Hwang H, Edgar JS, et al. Salivary α-synuclein and DJ-1: potential biomarkers for Parkinson’s disease. Brain. 2011;134:e178-e178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Vivacqua G, Suppa A, Mancinelli R, et al. Salivary alpha-synuclein in the diagnosis of Parkinson’s disease and progressive supranuclear palsy. Parkinsonism Relat Disord. 2019;63:143-148. [DOI] [PubMed] [Google Scholar]
- 197. Hansson O, Hall S, Ohrfelt A, et al. Levels of cerebrospinal fluid α-synuclein oligomers are increased in Parkinson’s disease with dementia and dementia with Lewy bodies compared to Alzheimer’s disease. Alzheimers Res Ther. 2014;6:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Parnetti L, Chiasserini D, Persichetti E, et al. Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in Parkinson’s disease. Mov Disord. 2014;29:1019-1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. El-Agnaf OM, Salem SA, Paleologou KE, et al. Detection of oligomeric forms of α-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006;20:419-425. [DOI] [PubMed] [Google Scholar]
- 200. Williams SM, Schulz P, Sierks MR. Oligomeric α-synuclein and β-amyloid variants as potential biomarkers for Parkinson’s and Alzheimer’s diseases. Eur J Neurosci. 2016;43:3-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Wang X, Yu S, Li F, Feng T. Detection of α-synuclein oligomers in red blood cells as a potential biomarker of Parkinson’s disease. Neurosc Lett. 2015;599:115-119. [DOI] [PubMed] [Google Scholar]
- 202. Pchelina S, Emelyanov A, Baydakova G, et al. Oligomeric α-synuclein and glucocerebrosidase activity levels in GBA-associated Parkinson’s disease. Neurosci Lett. 2017;636:70-76. [DOI] [PubMed] [Google Scholar]
- 203. Foulds PG, Mitchell JD, Parker A, et al. 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. [DOI] [PubMed] [Google Scholar]
- 204. Park MJ, Cheon S-M, Bae H-R, Kim S-H, Kim JW. Elevated levels of α-synuclein oligomer in the cerebrospinal fluid of drug-naïve patients with Parkinson’s disease. J Clin Neurol. 2011;7:215-222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Vivacqua G, Latorre A, Suppa A, et al. Abnormal salivary total and oligomeric alpha-synuclein in Parkinson’s disease. PLoS ONE. 2016;11:e0151156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Fujiwara H, Hasegawa M, Dohmae N, et al. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat Cell Biol. 2002;4:160-164. [DOI] [PubMed] [Google Scholar]
- 207. Majbour NK, Vaikath NN, van Dijk KD, et al. Oligomeric and phosphorylated alpha-synuclein as potential CSF biomarkers for Parkinson’s disease. Mol Neurodegener. 2016;11:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Shahnawaz M, Tokuda T, Waragai M, et al. Development of a biochemical diagnosis of Parkinson disease by detection of α-synuclein misfolded aggregates in cerebrospinal fluid. JAMA Neurol. 2017;74:163-172. [DOI] [PubMed] [Google Scholar]
- 209. Groveman BR, Orrù CD, Hughson AG, et al. Rapid and ultra-sensitive quantitation of disease-associated α-synuclein seeds in brain and cerebrospinal fluid by αSyn RT-QuIC. Acta Neuropathol Commun. 2018;6:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Fairfoul G, McGuire LI, Pal S, et al. Alpha-synuclein RT-Qu IC in the CSF of patients with alpha-synucleinopathies. Ann Clin Transl Neurol. 2016;3:812-818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Represa A, Deloulme JC, Sensenbrenner M, Ben-Ari Y, Baudier J. Neurogranin: immunocytochemical localization of a brain-specific protein kinase C substrate. J Neurosci. 1990;10:3782-3792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Gerendasy DD, Sutcliffe JG. RC3/neurogranin, a postsynaptic calpacitin for setting the response threshold to calcium influxes. Mol Neurobiol. 1997;15:131-163. [DOI] [PubMed] [Google Scholar]
- 213. Prichard L, Deloulme JC, Storm DR. Interactions between neurogranin and calmodulin in vivo. J Biol Chem. 1999;274:7689-7694. [DOI] [PubMed] [Google Scholar]
- 214. Domínguez-González I, Vázquez-Cuesta SN, Algaba A, Díez-Guerra FJ. Neurogranin binds to phosphatidic acid and associates to cellular membranes. Biochem J. 2007;404:31-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Huang K-P, Huang FL, Jäger T, Li J, Reymann KG, Balschun D. Neurogranin/RC3 enhances long-term potentiation and learning by promoting calcium-mediated signaling. J Neurosci. 2004;24:10660-10669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Thorsell A, Bjerke M, Gobom J, et al. Neurogranin in cerebrospinal fluid as a marker of synaptic degeneration in Alzheimer’s disease. Brain Res. 2010;1362:13-22. [DOI] [PubMed] [Google Scholar]
- 217. Kvartsberg H, Duits FH, Ingelsson M, et al. Cerebrospinal fluid levels of the synaptic protein neurogranin correlates with cognitive decline in prodromal Alzheimer’s disease. Alzheimers Dement. 2014;11:1180-1190. [DOI] [PubMed] [Google Scholar]
- 218. Kester MI, Teunissen CE, Crimmins DL, et al. Neurogranin as a cerebrospinal fluid biomarker for synaptic loss in symptomatic Alzheimer disease. JAMA Neurol. 2015;72:1275-1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Portelius E, Zetterberg H, Skillbäck T, et al. Cerebrospinal fluid neurogranin: relation to cognition and neurodegeneration in Alzheimer’s disease. Brain. 2015;138:3373-3385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. De Vos A, Jacobs D, Struyfs H, et al. C-terminal neurogranin is increased in cerebrospinal fluid but unchanged in plasma in Alzheimer’s disease. Alzheimers Dement. 2015;11:1461-1469. [DOI] [PubMed] [Google Scholar]
- 221. Tarawneh R, D’Angelo G, Crimmins D, et al. Diagnostic and prognostic utility of the synaptic marker neurogranin in Alzheimer disease. JAMA Neurol. 2016;73:561-571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Sanfilippo C, Forlenza O, Zetterberg H, Blennow K. Increased neurogranin concentrations in cerebrospinal fluid of Alzheimer’s disease and in mild cognitive impairment due to AD. J Neural Transm (Vienna). 2016;123:1443-1447. [DOI] [PubMed] [Google Scholar]
- 223. Lista S, Toschi N, Baldacci F, et al. Cerebrospinal fluid neurogranin as a biomarker of neurodegenerative diseases: a cross-sectional study. J Alzheimers Dis. 2017;59:1327-1334. [DOI] [PubMed] [Google Scholar]
- 224. Sutphen CL, McCue L, Herries EM, et al. Longitudinal decreases in multiple cerebrospinal fluid biomarkers of neuronal injury in symptomatic late onset Alzheimer’s disease. Alzheimers Dement. 2018;14:869-879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Blennow K, Diaz-Lucena D, Zetterberg H, et al. CSF neurogranin as a neuronal damage marker in CJD: a comparative study with AD. J Neurol Neurosurg Psychiatry. 2019;90:846-853. [DOI] [PubMed] [Google Scholar]
- 226. Wellington H, Paterson RW, Portelius E, et al. Increased CSF neurogranin concentration is specific to Alzheimer disease. Neurology. 2016;86:829-835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Portelius E, Olsson B, Höglund K, et al. Cerebrospinal fluid neurogranin concentration in neurodegeneration: relation to clinical phenotypes and neuropathology. Acta Neuropathol. 2018;136:363-376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Becker B, Nazir FH, Brinkmalm G, et al. Alzheimer-associated cerebrospinal fluid fragments of neurogranin are generated by Calpain-1 and prolyl endopeptidase. Molecular Neurodegeneration. 2018;13:1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Willemse EAJ, De Vos A, Herries EM, et al. Neurogranin as cerebrospinal fluid biomarker for Alzheimer disease: an assay comparison study. Clin Chem. 2018;64:927-937. [DOI] [PubMed] [Google Scholar]
- 230. Wesenhagen KEJ, Teunissen CE, Visser PJ, Tijms BM. Cerebrospinal fluid proteomics and biological heterogeneity in Alzheimer’s disease: a literature review. Crit Rev Clin Lab Sci. 2020;57:86-98. [DOI] [PubMed] [Google Scholar]
- 231. Brinkmalm G, Sjodin S, Simonsen AH, et al. A parallel reaction monitoring mass spectrometric method for analysis of potential CSF biomarkers for Alzheimer’s disease. Proteomics Clin Appl. 2018;12. doi: 10.1002/prca.201700131. [DOI] [PubMed] [Google Scholar]
- 232. Lleó A, Núñez-Llaves R, Alcolea D, et al. Changes in synaptic proteins precede neurodegeneration markers in preclinical Alzheimer’s disease cerebrospinal fluid. Mol Cell Proteomics. 2019;18:546-560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Xu D, Hopf C, Reddy R, et al. Narp and NP1 form heterocomplexes that function in developmental and activity-dependent synaptic plasticity. Neuron. 2003;39:513-528. [DOI] [PubMed] [Google Scholar]
- 234. Lee S-J, Wei M, Zhang C, et al. Presynaptic neuronal pentraxin receptor organizes excitatory and inhibitory synapses. J Neurosci. 2017;37:1062-1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Soldan A, Moghekar A, Walker KA, et al. Resting-state functional connectivity is associated with cerebrospinal fluid levels of the synaptic protein NPTX2 in non-demented older adults. Front Aging Neurosci. 2019;11:132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Spellman DS, Wildsmith KR, Honigberg LA, et al. Development and evaluation of a multiplexed mass spectrometry based assay for measuring candidate peptide biomarkers in Alzheimer’s Disease Neuroimaging Initiative (ADNI) CSF. Proteomics Clin Appl. 2015;9:715-731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Begcevic I, Tsolaki M, Brinc D, et al. Neuronal pentraxin receptor-1 is a new cerebrospinal fluid biomarker of Alzheimer’s disease progression. F1000Res. 2018;7:1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Wildsmith KR, Schauer SP, Smith AM, et al. Identification of longitudinally dynamic biomarkers in Alzheimer’s disease cerebrospinal fluid by targeted proteomics. Mol Neurodegener. 2014;9:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Llano DA, Bundela S, Mudar RA, Devanarayan V, Alzheimer’s Disease Neuroimaging Initiative (ADNI). A multivariate predictive modeling approach reveals a novel CSF peptide signature for both Alzheimer’s disease state classification and for predicting future disease progression. PLoS ONE. 2017;12:e0182098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Lim B, Tsolaki M, Soosaipillai A, et al. Liquid biopsy of cerebrospinal fluid identifies neuronal pentraxin receptor (NPTXR) as a biomarker of progression of Alzheimer’s disease. Clin Chem Lab Med. 2019;57:1875-1881. [DOI] [PubMed] [Google Scholar]
- 241. Lim B, Sando SB, Grøntvedt GR, Bråthen G, Diamandis EP. Cerebrospinal fluid neuronal pentraxin receptor as a biomarker of long-term progression of Alzheimer’s disease: a 24-month follow-up study. Neurobiol Aging. 2020;93:97.e1-97e7. [DOI] [PubMed] [Google Scholar]
- 242. Xiao MF, Xu D, Craig MT, et al. NPTX2 and cognitive dysfunction in Alzheimer’s disease. eLife. 2017;6:e23798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Swanson A, Willette AA, Alzheimer’s Disease Neuroimaging Initiative. Neuronal Pentraxin 2 predicts medial temporal atrophy and memory decline across the Alzheimer’s disease spectrum. Brain Behav Immun. 2016;58:201-208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Kroksveen A, Guldbrandsen A, Vedeler C, Myhr K, Opsahl J, Berven F. Cerebrospinal fluid proteome comparison between multiple sclerosis patients and controls. Acta Neurol Scand Suppl. 2012;195:90-96. [DOI] [PubMed] [Google Scholar]
- 245. van der Ende EL, Meeter LH, Stingl C, et al. Novel CSF biomarkers in genetic frontotemporal dementia identified by proteomics. Ann Clin Transl Neurol. 2019;6:698-707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Magdalinou N, Noyce A, Pinto R, et al. Identification of candidate cerebrospinal fluid biomarkers in parkinsonism using quantitative proteomics. Parkinsonism Relat Disord. 2017;37:65-71. [DOI] [PubMed] [Google Scholar]
- 247. Finnema SJ, Nabulsi NB, Eid T, et al. Imaging synaptic density in the living human brain. Sci Transl Med. 2016;8:348ra96. [DOI] [PubMed] [Google Scholar]
- 248. Bartholome O, Van den Ackerveken P, Sánchez Gil J, et al. Puzzling out synaptic vesicle 2 family members functions. Front Mol Neurosci. 2017;10:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. D’Alessandro R, Meldolesi J. Expression and function of the dense-core vesicle membranes are governed by the transcription repressor REST. FEBS Lett. 2013;587:1915-1922. [DOI] [PubMed] [Google Scholar]
- 250. Tanner VA, Ploug T, Tao-Cheng JH. Subcellular localization of SV2 and other secretory vesicle components in PC12 cells by an efficient method of preembedding EM immunocytochemistry for cell cultures. J Histochem Cytochem. 1996;44:1481-1488. [DOI] [PubMed] [Google Scholar]
- 251. Bajjalieh SM, Frantz GD, Weimann JM, Mcconnell SK, Scheller RH. Differential expression of synaptic vesicle protein 2 (SV2) isoforms. J Neuro. 1994;14:5223-5235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Crowder KM, Gunther JM, Jones TA, et al. Abnormal neurotransmission in mice lacking synaptic vesicle protein 2A (SV2A). Proc Natl Acad U S A. 1999;96:15268-15273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Nowack A, Yao J, Custer KL, Bajjalieh SM. SV2 regulates neurotransmitter release via multiple mechanisms. Am J Physiol Cell Physiol. 2010;299:C960-C967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Yao J, Nowack A, Kensel-Hammes P, Gardner RG, Bajjalieh SM. Cotrafficking of SV2 and synaptotagmin at the synapse. J Neurosci. 2010;30:5569-5578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Chen M-K, Mecca AP, Naganawa M, et al. Assessing synaptic density in Alzheimer disease with synaptic vesicle glycoprotein 2A positron emission tomographic imaging. JAMA Neurol. 2018;75:1215-1224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Robinson JL, Molina-Porcel L, Corrada MM, et al. Perforant path synaptic loss correlates with cognitive impairment and Alzheimer’s disease in the oldest-old. Brain. 2014;137:2578-2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Matuskey D, Tinaz S, Wilcox KC, et al. Synaptic changes in Parkinson disease assessed with in vivo imaging. Ann Neurol. 2020;87:329-338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Holland N, Jones PS, Savulich G, et al. Reduced synaptic density in progressive supranuclear palsy and corticobasal syndrome, revealed by [11C] UCB-J PET. medrxiv. 2020. https://www.medrxiv.org/content/10.1101/2020.01.24.20018697v2.
- 259. Ashton NJ, Höglund K, Leuzy A, et al. Cerebrospinal fluid synaptic vesicle glycoprotein 2A in Alzheimer's disease. Alzheimers Dement. 2019;15:P545. [Google Scholar]
- 260. Cho RW, Park JM, Wolff SB, et al. mGluR1/5-dependent long-term depression requires the regulated ectodomain cleavage of neuronal pentraxin NPR by TACE. Neuron. 2008;57:858-871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Kadavath H, Hofele RV, Biernat J, et al. Tau stabilizes microtubules by binding at the interface between tubulin heterodimers. Proc Natl Acad Sci U S A. 2015;112:7501-7506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Veleri S, Punnakkal P, Dunbar GL, Maiti P. Molecular insights into the roles of rab proteins in intracellular dynamics and neurodegenerative diseases. Neuromolecular Med. 2018;20:18-36. [DOI] [PubMed] [Google Scholar]
- 263. Laguerre F, Anouar Y, Montero-Hadjadje M. Chromogranin A in the early steps of the neurosecretory pathway. IUBMB Life. 2020;72:524-532. [DOI] [PubMed] [Google Scholar]
- 264. Blennow K, Davidsson P, Wallin A, Ekman R. Chromogranin A in cerebrospinal fluid: a biochemical marker for synaptic degeneration in Alzheimer’s disease. Dementia. 1995;6:306-311. [DOI] [PubMed] [Google Scholar]
- 265. Chatterjee M, Del Campo M, Morrema THJ, et al. Contactin-2, a synaptic and axonal protein, is reduced in cerebrospinal fluid and brain tissue in Alzheimer’s disease. Alzheimers Res Ther. 2018;10:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Galvin JE, Uryu K, Lee VM-Y, Trojanowski JQ. Axon pathology in Parkinson’s disease and Lewy body dementia hippocampus contains α-, β-, and γ-synuclein. Proc Natl Acad Sci U S A. 1999;96:13450-13455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Oeckl P, Halbgebauer S, Anderl-Straub S, et al. Targeted mass spectrometry suggests beta-synuclein as synaptic blood marker in Alzheimer’s disease. J Proteome Research. 2020;19:1310-1318. [DOI] [PubMed] [Google Scholar]
- 268. Zhang J, Zhou Y. 14-3-3 proteins in glutamatergic synapses. Neural Plast. 2018;2018:8407609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Foote M, Zhou Y. 14-3-3 proteins in neurological disorders. Int J Biochem Mol Biol. 2012;3:152-164. [PMC free article] [PubMed] [Google Scholar]
- 270. Antonell A, Tort-Merino A, RÃ-os J, et al. Synaptic, axonal damage and inflammatory cerebrospinal fluid biomarkers in neurodegenerative dementias. Alzheimers Dement. 2020;16:262-272. [DOI] [PubMed] [Google Scholar]
- 271. Sze C-I, Troncoso JC, Kawas C, Mouton P, Price DL, Martin LJ. Loss of the presynaptic vesicle protein synaptophysin in hippocampus correlates with cognitive decline in Alzheimer disease. J Neuropathol Exp Neurol. 1997;56:933-944. [DOI] [PubMed] [Google Scholar]
- 272. Davidsson P, Blennow K. Neurochemical dissection of synaptic pathology in Alzheimer’s disease. Int Psychogeriatr. 1998;10:11-23. [DOI] [PubMed] [Google Scholar]
- 273. Schlaf G, Salje C, Wetter A, Stuertz K, Felgenhauer K, Mäder M. Determination of synapsin I and synaptophysin in body fluids by two- site enzyme-linked immunosorbent assays. J Immunol Methods. 1998;213:191-199. [DOI] [PubMed] [Google Scholar]
- 274. Goetzl EJ, Kapogiannis D, Schwartz JB, et al. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 2016;30:4141-4148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Manek R, Moghieb A, Yang Z, et al. Protein biomarkers and neuroproteomics characterization of microvesicles/exosomes from human cerebrospinal fluid following traumatic brain injury. Mol Neurobiol. 2018;55:6112-6128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Chiasserini D, van Weering JR, Piersma SR, et al. Proteomic analysis of cerebrospinal fluid extracellular vesicles: a comprehensive dataset. Journal of Proteomics. 2014;106:191-204. [DOI] [PubMed] [Google Scholar]
- 277. Goetzl EJ, Abner EL, Jicha GA, Kapogiannis D, Schwartz JB. Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer’s disease. FASEB J. 2018;32:888-893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Shi M, Liu C, Cook TJ, et al. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol. 2014;128:639-650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Bellingham SA, Guo B, Coleman B, Hill AF. Exosomes: vehicles for the transfer of toxic proteins associated with neurodegenerative diseases. Front Physiol. 2012;3:124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280. Janas AM, Sapoń K, Janas T, Stowell MH, Janas T. Exosomes and other extracellular vesicles in neural cells and neurodegenerative diseases. Biochim Biophys Acta. 2016;1858:1139-1151. [DOI] [PubMed] [Google Scholar]
- 281. Mollenhauer B, Cullen V, Kahn I, et al. Direct quantification of CSF α-synuclein by ELISA and first cross-sectional study in patients with neurodegeneration. Exp Neurol. 2008;213:315-325. [DOI] [PubMed] [Google Scholar]
- 282. Leinonen V, Menon LG, Carroll RS, et al. Cerebrospinal fluid biomarkers in idiopathic normal pressure hydrocephalus. Int J Alzheimers Dis. 2011;2011:312526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Sjögren M, Blomberg M, Jonsson M, et al. Neurofilament protein in cerebrospinal fluid: a marker of white matter changes. J Neurosci Res. 2001;66:510-516. [DOI] [PubMed] [Google Scholar]
- 284. Sjögren M, Rosengren L, Minthon L, Davidsson P, Blennow K, Wallin A. Cytoskeleton proteins in CSF distinguish frontotemporal dementia from AD. Neurology. 2000;54:1960-1964. [DOI] [PubMed] [Google Scholar]
- 285. Kušnierová P, Zeman D, Hradílek P, Čábal M, Zapletalová O. Neurofilament levels in patients with neurological diseases: a comparison of neurofilament light and heavy chain levels. J Clin Lab Anal. 2019;33:e22948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Kuhle J, Plattner K, Bestwick JP, et al. A comparative study of CSF neurofilament light and heavy chain protein in MS. Mult Scler. 2013;19:1597-1603. [DOI] [PubMed] [Google Scholar]
- 287. Weston PS, Poole T, Ryan NS, et al. Serum neurofilament light in familial Alzheimer disease: a marker of early neurodegeneration. Neurology. 2017;89:2167-2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Preische O, Schultz SA, Apel A, et al. Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer’s disease. Nat Med. 2019;25:277-283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Schmitz M, Ebert E, Stoeck K, et al. Validation of 14-3-3 protein as a marker in sporadic Creutzfeldt-Jakob disease diagnostic. Mol Neurobiol. 2016;53:2189-2199. [DOI] [PubMed] [Google Scholar]
- 290. Lauterborn JC, Cox CD, Chan SW, Vanderklish PW, Lynch G, Gall CM. Synaptic actin stabilization protein loss in Down syndrome and Alzheimer disease. Brain Pathol. 2019;30:319-331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. Keller R, Basta R, Salerno L, Elia M. Autism, epilepsy, and synaptopathies: a not rare association. Neurol Sci. 2017;38:1353-1361. [DOI] [PubMed] [Google Scholar]
- 292. Grant SG. Synaptopathies: diseases of the synaptome. Curr Opin Neurobiol. 2012;22:522-529. [DOI] [PubMed] [Google Scholar]
- 293. Scheff SW, Neltner JH, Nelson PT. Is synaptic loss a unique hallmark of Alzheimer’s disease? Biochemical Pharmacology. 2014;88:517-528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. de Wilde MC, Overk CR, Sijben JW, Masliah E. Meta-analysis of synaptic pathology in Alzheimer’s disease reveals selective molecular vesicular machinery vulnerability. Alzheimers Dement. 2016;12:633-644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295. Almeida CG, Tampellini D, Takahashi RH, et al. Beta-amyloid accumulation in APP mutant neurons reduces PSD-95 and GluR1 in synapses. Neurobiol Dis. 2005;20:187-198. [DOI] [PubMed] [Google Scholar]
- 296. Canas PM, Simoes AP, Rodrigues RJ, Cunha RA. Predominant loss of glutamatergic terminal markers in a beta-amyloid peptide model of Alzheimer’s disease. Neuropharmacology. 2014;76:51-56. [DOI] [PubMed] [Google Scholar]
- 297. Roselli F, Tirard M, Lu J, et al. Soluble beta-amyloid1-40 induces NMDA-dependent degradation of postsynaptic density-95 at glutamatergic synapses. J Neurosci. 2005;25:11061-11070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298. Ekman U, Ferreira D, Westman E. The A/T/N biomarker scheme and patterns of brain atrophy assessed in mild cognitive impairment. Sci Rep. 2018;8:8431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Ashton NJ, Hye A, Rajkumar AP, et al. An update on blood-based biomarkers for non-Alzheimer neurodegenerative disorders. Nat Rev Neurol. 2020;16:265-284. [DOI] [PubMed] [Google Scholar]
- 300. Ferretti MT, Martinkova J, Biskup E, et al. Sex and gender differences in Alzheimer’s disease: current challenges and implications for clinical practice: position paper of the Dementia and Cognitive Disorders Panel of the European Academy of Neurology. Eur J Neurol. 2020;27:928-943. [DOI] [PubMed] [Google Scholar]
- 301. DeFelipe J, Alonso-Nanclares L. The synapse: differences between men and women. In: Pfaff DW, Christen Y, eds. Multiple Origins of Sex Differences in Brain. Research and Perspectives in Endocrine Interactions. Berlin, Heidelberg: Springer; 2013:43-57. [Google Scholar]