Abstract
The culmination of many years of increasing research into the toxicity of tau aggregation in neurodegenerative disease has led to the consensus that soluble, oligomeric forms of tau are likely the most toxic entities in disease. While tauopathies overlap in the presence of tau pathology, each disease has a unique combination of symptoms and pathological features, however, most study into tau has grouped tau oligomers and studied them as a homogenous population. Established evidence from the prion field combined with the most recent tau and amyloidogenic protein research suggests that tau is a prion-like protein, capable of seeding the spread of pathology throughout the brain. Thus, it is likely that tau may also form prion-like strains or diverse conformational structures that may differ by disease and underlie some of the differences in symptoms and pathology in neurodegenerative tauopathies. The development of techniques and new technology for the detection of tau oligomeric strains may therefore lead to more efficacious diagnostic and treatment strategies for neurodegenerative disease.
Keywords: Tau, Amyloid, Oligomer, Neurodegeneration, Aggregation
Introduction
In the search for an effective therapeutic strategy against Alzheimer’s disease (AD), the most common cause of dementia worldwide, aggregated amyloid-β (Aβ) emerged as the main target of interest and has remained so for many years. However, as more and more clinical trials against the protein failed, researchers searched for a new and hopefully more effective route to treatment(Godyń et al., 2016). The pathological aggregation of the microtubule-associated protein tau and its subsequent deposition in neurofibrillary tangles (NFTs) are also defining histopathological features of AD that correlate better with symptoms than Aβ(Braak and Braak, 1995). Therefore, the study of tau has become an active field of study with a lot of promise(Šimić et al., 2016). Tau aggregation also plays a role in many other neurodegenerative disorders, collectively known as tauopathies, including Pick’s disease (PiD), progressive supranuclear palsy (PSP), dementia with Lewy bodies (DLB), Parkinson’s disease (PD), frontotemporal dementia (FTD), and corticobasal degeneration (CBD)(Ballatore et al., 2007, Gendron and Petrucelli, 2009). Although this large group of related disorders shares the common pathological hallmark of accumulated tau protein aggregates in the brain, they are diversified with a vast array of pathologies and symptom progressions.
NFTs may play a role in the pathophysiology of tauopathies, but little is known about the mechanisms underlying their formation and the role of tau intermediates soluble in physiological fluids formed between tau monomers and filaments. Hyperphosphorylation contributes to the formation of NFTs, which until recently were believed to trigger processes that lead to neuronal cell death(Andrade-Moraes et al., 2013). However, NFT accumulation alone may be insufficient to cause cell death, as neuropathological studies in AD patients suggest that neuronal loss and cognitive deficits precede NFT formation(Haroutunian et al., 2007). Supporting this, animal models show that tau oligomers appear prior to NFTs and contribute to learning and memory deficits and neuronal cell death, while NFTs are not associated with neuronal death(Lasagna-Reeves et al., 2011b, Cowan et al., 2012, Cook et al., 2015, Kim et al., 2016).
Despite increasing research on this topic, the mechanism by which tau induces toxicity and disease progression remains unknown. One of the largest barriers to conclusive mechanistic investigations of amyloidogenic and tau proteins is inconsistency in the protein species (oligomer, fibril, non-specifically defined aggregate, etc.) studied(Cowan and Mudher, 2013), an important consideration as the aggregation state of tau is critical to its function. Within the same aggregation state, tau exhibits conformational differences that could exert diverse downstream effects(Hyman, 2014, Sanders et al., 2014). Many parallels exist between prions and amyloidogenic proteins present in neurodegenerative disease, suggesting that tau and other aggregate-prone proteins may form prion-like conformers or strains that affect disease progression. Therefore, understanding the characteristics of different tau aggregates may be critical for the investigation of tau’s toxicity and may be responsible for the pathological and symptomatic differences underlying various tauopathies(Hyman, 2014).
Prion Structural Polymorphisms and Strains
One of the most puzzling phenomena in prion biology is the existence of prion strains, defined as infectious protein isolates that, when transmitted to identical hosts, induce distinct prion-disease phenotypes(Bessen, 1992, Bartz, 2000). The phenotypic traits include incubation times, histopathological lesion profiles, and specific neuronal target areas. These traits typically persist following serial transmissions, thereby stabilizing strain conformation(Bartz, 2000). The strain phenotypes might be encoded by the same aggregate assuming different conformations that have distinct properties, defined as “strains or conformers”. These conformations can also be manipulated(Legname et al., 2005, Tanaka et al., 2006). It has been demonstrated that multiple strains may exist in different brain regions in the same patient, and this is hypothesized to underlie the individual variability in the clinical manifestations of patients with Creutzfeldt-Jakob disease(Schoch, 2006). Prion strains can compete with one another, suggesting that less pathogenic prions could be used as a therapeutic agent against the most toxic strains(Morales et al., 2007). A recent study in the prion field showed that a single mutation leading to a particular prion strain led to complete protection against toxic misfolded prion protein due to an inability to propagate the toxic prion conformation(Asante et al., 2015). The strain phenomenon has recently been hypothesized to extend to many amyloidogenic proteins that form distinct conformations and can be classified as strains, including Aβ(Heilbronner G, 2013, Lu JX, 2013) and α-synuclein(Guo et al., 2013).
Amyloidogenic Protein Oligomeric Strains
Similarly to tau, many groups have reported that fibrils, once thought to be the most toxic species, are actually less toxic than intermediate Aβ aggregates (spherical oligomers and protofibrils)(Glabe and Kayed, 2006). There are a variety of Aβ oligomer conformations(Glabe, 2008) that can be produced by several pathways. Simple manipulation of conditions in which Aβ aggregates can alter conformation, highlighting the complexity of oligomer formation(Kayed et al., 2003, Glabe and Kayed, 2006, Kayed and Glabe, 2006, Kayed et al., 2007, Necula et al., 2007, Glabe, 2008, Mina et al., 2009). Several studies suggest that oligomeric species form via different mechanisms and exhibit different toxicities(Cizas et al., 2010, Zako, 2010, Kayed and Lasagna-Reeves, 2013). A recent study found that two different Aβ oligomers that differ in toxicity can be differentiated using conformation-specific antibodies(Liu et al., 2015). Aβ conformational differences have also been specifically correlated with more rapidly progressive AD(Cohen et al., 2015). Moreover, structurally distinct α-synuclein strains can lead to different behavioral phenotypes and levels of toxicity(Peelaerts et al., 2015) and may be differentiated using similar techniques used in the prion field, such as resistance to proteases(Roberts et al., 2015). Strains of α-synuclein derived from multiple systems atrophy (MSA) were recently shown to be capable of serial transmission in cell culture as well as to induce neuronal dysfunction following successive inoculations in mice(Prusiner et al., 2015, Woerman et al., 2015). Thus a variety of oligomeric species may be common to many proteinopathies and underpin evolution of disease, as well as distinct sub-types of disease.
Classes of Aggregated Tau
Due to the variety of diseases involving aggregated tau with distinct pathological features and symptomology, conformational differences in tau may be a crucial factor distinguishing tauopathies. Different conformers or strains may be present in one or many of the forms of aggregated tau. Soluble aggregation intermediates include protofibrils, annular protofibrils and oligomers, which have subtypes as well that form either en route to insoluble fibril formation or independently to fibrillization.
Oligomers
Tau in its native state exists as an unfolded monomer which has an important function stabilizing microtubules. Tau undergoes alternative splicing to form six different protein isoforms (Fig. 1A) present at varying levels during development, normal adulthood, and disease states. The isoform of tau has important implications for its structure as the number of microtubule binding repeats in the stable core of affects its aggregation stability and the unstructured N- and C-terminal domains may underlie much of the conformational diversity as they provide environmentally-dependent flexibility to the protein(Wegmann et al., 2013, Xu et al., 2016)(Fig. 1B). The unfolded state of monomeric tau, when released from microtubules allows intermolecular contacts and exposure of hydrophobic regions, which combined with a number of environmental factors can lead to the misfolding and aggregation of tau (Fig. 2). Multimeric tau species which form, known as tau oligomers, have repeatedly been found to induce neurotoxicity and cell death in neurodegenerative tauopathies, rather than the large fibrillar structures known as NFTs(Spires et al., 2006, Berger et al., 2007, Maeda et al., 2007, Kopeikina et al., 2011, Cowan and Mudher, 2013, Sahara et al., 2013, Cowan et al., 2015). We have shown that these dynamic tau aggregates are detected in human disease and can propagate through the brain, causing synaptic and mitochondrial dysfunction associated with memory deficits when administered to cognitively normal mice(Lasagna-Reeves et al., 2011a, Lasagna-Reeves et al., 2012b). Importantly, reducing levels of tau oligomers with oligomer-specific antibodies leads to protection against behavioral deficits and tau pathology in multiple mouse models(Castillo-Carranza et al., 2014a, Castillo-Carranza et al., 2014b, Castillo-Carranza et al., 2015).
Fig. 1. Alternative splicing leads to the formation of six different tau protein isoforms.
(A) Alternative splicing of tau and tau isoform makeup may alter protein folding and aggregate conformation. Tau forms six different isoforms, three of which have three repeats in the microtubule binding domain (3R0N, 3R1N, 3R2N) and three of which have four repeats (4R0N, 4R1N, 4R2N). (B) Tau isoforms may lead to differential stability and structure in tau oligomers depending upon the rigidity of the structure. Modifications and environmental conditions can alter the unstructured C- and N-termini surrounding the stable, protease-resistant core.
Fig. 2. Tau oligomers are the most toxic species in disease.
A number of complex and inter-related factors, including alterations in post-translational modifications, truncation alternative splicing, mutations, co-factors, chaperones, oxidative stress and seeds (Aβ, α-synuclein) can lead to the formation of soluble, intermediate tau aggregates—tau oligomers. Tau oligomers may go on to form stable filaments, however, these highly dynamic structures appear to be far more toxic than tau fibrils and may lead to the spread of pathology and a number of downstream toxic effects. Thus, tau oligomers represent a bottleneck in the process of tau toxicity in disease and a likely important therapeutic target.
Protofibrils
Protofibrils are intermediate, metastable species that form en route to fibrillization. Protofibrils share some structural similarities with mature fibrils, including a rope-like fibrillar structure detectable by EM and AFM and stable hydrogen bonding. However, protofibrils are inherently more dynamic and unstable and do not show high response to thioflavin T(Williams et al., 2005a), which recognizes the β-sheet structure characteristic of amyloid fibrils. Protofibrils can be detected with oligomer-specific conformational antibodies, indicating their similar structure (Kayed et al., 2003). Similarly to oligomers, these soluble intermediates have been shown to be more toxic and relevant to disease pathogenesis than the larger, more stable fibrillar structures and NFTs (Harper et al., 1997, Walsh et al., 1997, Habicht et al., 2007, Habicht et al., 2008, Rijal Upadhaya et al., 2012)
Annular Protofibrils
The formation of pore-like structures, known as annular protofibrils, has been known for amyloid proteins, including amyloid-β and α-synuclein for years(Hafner et al., 2001, Kril et al., 2002, Lasagna-Reeves et al., 2011c). Recently, the formation of tau annular protofibrils was shown in vitro and in human disease tissue(Lasagna-Reeves et al., 2014). Results suggest that tau oligomers can go on to form annular protofibrils by a specific off-pathway mechanism from fibril formation. These pore-like structures are believed to disrupt permeability of the cell membrane, leading to non-specific ion leakage.
Tau Fibrils
Paired helical filaments (PHF) and straight filaments (SF) that make up NFTs are comprised of hyperphosphorylated tau(Grundke-Iqbal et al., 1986, Lee et al., 1991) in a β-sheet conformation(von Bergen et al., 2005). Differences in NFT location and structure exist among various tauopathies. In a study by Clavaguera et al., brain homogenate derived from various tauopathies was injected into the brains of ALZ17 mice, which express non-mutated human tau, and this induced the deposition and spread of characteristic tau aggregates from each disease(Clavaguera et al., 2013). Recently, Boluda et al. extended these results, showing that at much earlier time points than the original study, CBD and AD-specific pathology propagates in ALZ17 mice following brain homogenate injection, with distinct cell specificity(Boluda et al., 2015).
While this is strong evidence for the existence of tau strains, it is critically important to accurately identify the species in total brain homogenate responsible for seeding the spread of unique tau pathologies. In addition to isoform composition, research suggests that disulfide linkages impact fibril structure(Furukawa et al., 2011). Sanders et al. (2014) reported that conformationally distinct tau fibrils generated from the tau repeat domain propagate in cells over multiple passages. Additionally, when injected bilaterally in the hippocampi of P301S mice, different strains are propagated over multiple generations and can be differentiated by their biochemical characteristics(Sanders et al., 2014). However, fibrillar strains were derived from tau fragments, which may not accurately reflect natural conditions in human disease and as no behavior was completed, toxicity in vivo is difficult to assess from this study(Sanders et al., 2014). Nonetheless this study provided good evidence for the idea that the conformational diversity of tau strains dictates its toxic potential and transmissibility. In addition, expressing specific point mutations in truncated tau also leads to diversification of fibrillar structure(Meyer et al., 2014).
Thus it is of great interest to determine whether toxic tau oligomers form distinct strains that differ by disease. Similarities between tau and other amyloidogenic proteins with unique conformational strains suggest that this is likely the case (Fig. 3).
Fig. 3. Hypothetical models for the formation of tau oligomeric conformers.
(A) Risk factors cause tau misfolding and lead to the formation of conformationally distinct misfolded monomers that aggregate into different tau oligomeric strains. (B) Misfolded tau induces the formation of a flexible, dynamic, master oligomeric conformer that transforms into different oligomeric strains depending on less-defined risk factors, such as changes to cell autonomous and non-autonomous factors. (C) Conformational differences in tau oligomeric strains could lead to differential seeding of the spread of strains in different brain regions and cell populations.
Other Potentially Toxic Species
Misfolded tau monomer is another possibly toxic species in tauopathies. While most studies show that tau monomer is non-toxic and incapable of propagating tau pathology(Lasagna-Reeves et al., 2012a, Wu et al., 2013), an abnormal, higher molecular weight form of tau monomer known as PHF-tau or A68 has been shown to exhibit some potentially toxic characteristics(Liu, 1993). The 68 kD tau is found in higher proportions in tau aggregates in disease brains and is more highly phosphorylated than native tau monomer(Bramblett et al., 1993, Shin et al., 1993, Miller, 1999). Truncated tau may also be a toxic species of interest. Fragmentation of tau has also been shown to be affected in disease states, led to increased aggregation in vitro, and increased toxicity(Zilka et al., 2006, De Strooper, 2010, Reifert et al., 2011).
Tau Oligomeric Strains
Collectively the evidence suggests that dynamic tau intermediate soluble aggregates (tau oligomers) are the toxic tau entities and initiators of tau pathology and propagation. The formation of other amyloidogenic protein oligomeric strains and evidence for tau fibrillar strains suggests that tau oligomers may exist in different conformationally distinct conformers (strains) that play a critical role in disease phenotypes. One pre-fibrillar tau oligomer species that has been characterized and appears to have differential structure and toxicity is the granular tau oligomer(Maeda et al., 2006, Maeda et al., 2007), reviewed by Cowan and Mudher(Cowan and Mudher, 2013). This is a 40mer structure identified in AD brains and believed to be a precursor of tangles. However, it does not appear to be as clearly toxic as other oligomeric tau species since it can be found in animal models in the absence of toxicity(Cowan et al., 2015), similarly to the fibrillar Aβ oligomer strain that was found to be less toxic than other conformational states(Liu et al., 2015). The reason for the lowered toxicity could be due to a lack of beta pleated sheet conformation in granular tau oligomers. This illustrates the fact that tau aggregates, whether small or intermediate in size, vary in constituent tau composition and conformation and this may play a role in determining their toxicity. Thus, the elucidation of the different tau oligomeric conformers may reveal new targets for novel therapeutic strategies tailored for different neurodegenerative tauopathies and perhaps could be used to screen the best candidates and experimental drugs.
Tau Aggregation as a Secondary Amyloidosis
The critical role of amyloidogenic oligomers has been revealed for multiple neurodegenerative diseases(Walsh and Selkoe, 2004). Recently published works support the idea that prefibrillar soluble oligomers of amyloidogenic proteins, including α-synuclein and amyloid-beta (Aβ) are more toxic than insoluble Lewy bodies and senile plaques(Klein et al., 2001, Walsh et al., 2002, Gispert et al., 2003, Kayed et al., 2003, Lesne et al., 2006, Walsh and Selkoe, 2007, Danzer et al., 2009, Tsika et al., 2010). Pathogenic proteins involved in different neurological disorders often interact with one another and modulate downstream activities that disrupt their normal function. Although Alzheimer’s disease and Parkinson’s disease have unique pathological features, there is considerable overlap between these two disorders. The effects of Aβ aggregation on tau misfolding and aggregation are well-established(Hardy, 2002, Oddo et al., 2006, Clinton et al., 2010, Bloom, 2014). It is also well-established that aggregated Aβ is an important contributor to tau phosphorylation and aggregation in animal models and cell cultures. In primary neuronal culture, Aβ is capable of inducing tau phosphorylation(Busciglio et al., 1995). Aβ42 fibrils induced formation of NFTs in P301L tau transgenic mice(Gotz et al., 2001), and pre-aggregated Aβ42 induced tau paired helical filament (PHF) formation in cells over-expressing human tau(Ferrari et al., 2003, Pennanen and Gotz, 2005). These experiments were performed with aggregated Aβ, which implies that they contain different prefibrillar and fibrillar Aβ aggregates. Recently, it has been shown that soluble Aβ oligomers trigger tau aggregation in vitro and in vivo(Chabrier et al., 2012). We previously demonstrated that Aβ and α-synuclein oligomers prepared in vitro are capable of inducing toxic tau oligomer formation(Lasagna-Reeves et al., 2010), targeting tau oligomers in an Aβ precursor protein (APP) overexpression mouse model also leads to a decrease in toxic Aβ aggregates and a genome-wide association study reported genetic interactions between tau and α-synuclein in Parkinson’s disease pathogenesis(Simon-Sanchez et al., 2009). Moreover, fibrillar α-synuclein has been shown to induce tau tangle formation in vitro(Waxman and Giasson, 2011) and accelerate tau phosphorylation, further enhancing its likelihood to form aggregates(Jensen et al., 1999). Recently two different α-synuclein fibrillar seeds have been identified with varying capacities of causing tau aggregation both in neuronal cultures and in vivo(Guo et al., 2013). A study also provided evidence that α-synuclein induces fibrillar tau formation and found that both tau and α-synuclein exert synergistic effects on each other leading to the formation of fibrillar amyloid structure(Giasson et al., 2003). Although insoluble forms of α-synuclein and tau proteins have been observed to co-exist in DLB patients(Iseki et al., 2002), our recent report was the first published evidence that these proteins occur together within oligomeric organizations that further perpetuate their aggregation(Sengupta et al., 2015).
Collectively this illustrates that the complexity of neurodegenerative tauopathies is likely to be partially dependent upon interactions between other proteins, particularly other amyloids. Cross-seeding between aggregant proteins may induce the formation of diverse tau conformers differing between diseases with mixed protein pathologies. However, in addition to the common mixed pathology disorders—AD, PD, and DLB—phenotypes and pathologies also differ between tauopathies lacking aggregation of other amyloidogenic proteins. Therefore, other mechanisms must be at play that are independent of other aggregant proteins.
Tau Modifications
Modifications to tau take place in the form of alternative splicing, mutations, post-translational modifications, and truncation. These changes could alter the folding of the protein and lead to the formation of different conformations. In support of this idea is the fact that tauopathies are associated with different tau isoform compositions. For example, PSP and CBD specifically display primarily 4R tau pathology and 3R tau aggregation, respectively, while PiD and AD display both 3R and 4R aggregates. Studies have found that 3R tau forms mainly twisted PHFs, while 4R tau fibrils are mainly SFs in vitro(Goedert, 1996, Barghorn and Mandelkow, 2002). However, the in vivo fibrillar conformations of isoform-specific aggregates in PiD and CBD vary(Munoz-Garcia, 1984, Ksiezak-Reding, 1994). Another likely cause of conformational differences in disease proteins is the presence of genetic mutations. While tau mutations are not associated with AD or PD, they do induce other tauopathies. The identification of around 40 tau mutations that cause familial dementia, such as FTD and Parkinsonism linked to chromosome 17 (FTDP-17) demonstrated that tau alone could cause neurodegeneration(Goedert and Jakes, 2005). However, the mechanisms by which tau mutations cause neurological disorders are still unclear, though the effect of mutations on tau aggregation and fibril formation has been studied extensively. While tau protein will not spontaneously aggregate in vitro without the addition of polyanionic cofactors, tau fragments expressing point mutations, P301L and ΔK280, rapidly aggregate due to an increase in the shift towards β-sheet structure(von Bergen et al., 2001). Multiple mutations have been identified in the microtubule binding repeat region (P301S, P301L, and P301T), which highlights its importance in aggregation. Additionally, in a study on the effect of mutations on heparin-induced fibrillar assembly, P301L and P301S mutations led to the largest increase in filament formation. G272V also increased filament formation in both 3R and 4R tau, while V337M had a small stimulatory effect only in 3R tau isoforms(Goedert et al., 1999). Studies carried out both in vitro and in vivo suggest that different mutations may have varied phenotypic and pathogenic effects. The P301S mutation is associated with early onset of clinical symptoms of FTDP-17, reduced microtubule assembly, and increased filament assembly. P301S mice show motor symptoms, including severe partial paralysis of the hindlimbs as well as hyperphosphorylated tau aggregates in the brain and spinal cord, but no signs of apoptosis (Allen et al., 2002). On the other hand, P301L mice have increased levels of apoptosis(Götz et al., 2001, Ho et al., 2001). These mice also display motor deficits and NFTs in the brain and spinal cord, as well as deposits in glial cells(Lewis J, 2000, Götz et al., 2001). Meanwhile, the R406W mutation associated with FTDP-17 induces similar clinical symptoms and neuropathology seen in Alzheimer’s disease. Mice expressing the mutation develop NFTs of similar morphology to AD inclusions in the forebrain and cognitive deficits, but no sensorimotor symptoms(Tatebayashi et al., 2002). Moreover, a recent study revealed that mutations give rise to varied tau fibrillar strains(Meyer et al., 2014). Phosphorylated tau is well-known to be altered in disease and to affect aggregation. However, the relationship between phosphorylation state and toxicity is complex, with different sites leading to widely varying effects, as was illustrated recently in a Drosophila model of tauopathy(Papanikolopoulou and Skoulakis, 2015). In fact, elevated levels of tau phosphatase has been shown to increase toxicity of tau in the extracellular space(Díaz-Hernández et al., 2010). Moreover in a Drosophila model of tauopathy, reduced tau phosphorylation was associated with rescue of phenotype but increased oligomerisation of tau(Cowan et al., 2015). Tau has also been shown to be altered by a number of other post-translational modifications including acetylation, glycation, and others(Gerson et al., 2014), suggesting that this wide variability could have an effect on tau oligomer conformation. Tau has also been shown to form disease-specific fragments that enhance its ability to seed the formation of oligomers and spread through the extracellular space(Wang et al., 2009, Kanmert et al., 2015, Matsumoto et al., 2015). These results suggest that tau modifications could be involved in the formation of unique oligomeric tau conformations that result in different disease phenotypes.
Cell and organelle specificity
Regional and cellular differences in accumulation of tau aggregates between neurodegenerative diseases have been long known and used to differentiate disorders, even prior to the proposal of the prion-like attributes of tau and other aggregant proteins. Alzheimer’s disease is characterized by the presence of flame-shaped NFTs in neurons and rarely glial inclusions. In Pick’s disease, tau aggregates are primarily found in spherical Pick bodies in neurons, but can also be detected in lower levels in astrocytes and oligodendrocytes, while in both PSP and CBD there is widespread deposition of astrocytic and oligodendroglial tau aggregates(Yoshida, 2006). Moreover, we found that while multiple tauopathies contain tau annular protofibrils, cellular specificity of their formation differed by disease, suggesting the presence of disease and cell-specific conformations(Lasagna-Reeves et al., 2014). Additionally, alterations to the intracellular localization of tau may correlate with disease. While tau in its native state is commonly located in the axonal cytoskeleton, recent studies suggest that it also has a nuclear role, binding and protecting DNA and RNA under oxidative stress conditions that also lead to the formation of toxic tau oligomers(Sultan et al., 2011, Violet et al., 2014). In this environment, functional tau translocates from the cytosol to the nucleus, however tau oligomer formation appears to both inhibit the movement of protective tau to the nucleus and lead to the buildup of nuclear tau aggregates(Violet et al., 2015). Tau may also differentially interact with RNA in disease states in the endoplasmic reticulum (ER) as tau has been shown to be associated with ribosomal and other ER proteins(Meier, 2015). The redistribution of tau protein may also underlie synaptic dysfunction seen in tauopathies as tau aggregates have been found in higher proportions in the somatodendritic compartment and can be specifically detected at the pre- and post-synaptic densities in disease states(Meyer et al., 2014).
Spread of tau oligomeric strains
The ability of amyloid and tau fibrils to seed soluble monomers and convert them to aggregates capable of propagating has been known for more than 20 years and is well standardized and documented(Jarrett et al., 1993, Kelly, 2000, Margittai and Langen, 2004, O’Nuallain et al., 2004, Siddiqua and Margittai, 2010, Dinkel et al., 2011). Recently, our lab and others have shown that amyloid oligomers also can seed and propagate in vitro(Kayed et al., Lee et al., 2011). Moreover, we discovered that tau oligomers prepared from recombinant tau can seed the aggregation of tau in vitro(Lasagna-Reeves et al., 2010). It is hypothesized that brain-derived tau oligomers seed and propagate by a mechanism known as oligomer-nucleated conformational induction. Unlike the seeding mechanism proposed for tau fibrils prepared in vitro—template-assisted growth(Margittai and Langen, 2004, Congdon et al., 2008)—this mechanism excludes the addition of tau monomer to the nucleus/template, stating instead that all monomers will assemble into oligomers before fibril formation. In this mechanism, different conformational strains of tau oligomers would propagate rapidly, seeding endogenous monomer to oligomerize prior to forming stable fibrillar structures.
There is increasing interest in understanding the spread of disease from the early stages of tauopathies. In the specific case of AD, NFTs progressively spread throughout the brain in an anatomically stereotypical manner(Braak and Braak, 1991, Delacourte et al., 2002). Based on these and several other studies, it has been postulated that tau proteins spread via a prion-like mechanism(Clavaguera et al., 2009, Brundin et al., 2010, Frost and Diamond, 2010). Clavaguera and coworkers support this concept by demonstrating that intracerebral injections of brain extracts from mice expressing human P301S tau induce the formation and spread of tau aggregates. The pathology spreads from the injection site to neighboring brain regions 15 months post-injection in mice transgenic for human wild type tau (ALZ17 mouse model)(Clavaguera et al., 2009) and 12 months post-injection in wild-type mice. The observation that some areas of the human brain show evidence of neurodegeneration prior to the formation of NFTs and clinical diagnosis(Kril et al., 2002) suggests that tau oligomers could be responsible for neuronal death and the spread of tau pathology. Up to this point, all studies investigating the propagation of tau in cells and in vivo have used heterogeneous populations of aggregated tau; therefore, whether tau oligomers—as opposed to other species—play a leading role in tau aggregation and propagation has been difficult to ascertain. A recent publication by Mathias Jucker and Larry Walker demonstrated that soluble Aβ assemblies (oligomers) are the most potent amyloid-inducing factors, 50-fold stronger than large fibrillar aggregates(Langer et al., 2011).
These results combined with evidence for the existence of different tau oligomeric strains suggest that varying regional distribution and time to acquire disease may be due to differences in the ability of tau conformers to seed and spread through the brain. Within the same disease, some strains may correlate with certain disease stages. There may be diversity by brain region as well as by cell type as different environmental conditions including pH(Sneideris et al., 2015, Verasdonck et al., 2015) and temperature(Tanaka et al., 2006) could lead to conformational changes. The impact of interactions with chaperones, as well as post-translational modifications are also important areas of exploration as the formation of amyloid strains has been shown to depend on variability of chaperone interaction(Sporn and Hines, 2015) and tau is known to interact with a number of different chaperones that affect its aggregation(Petrucelli et al., 2004, Dickey et al., 2006). Additionally, differences in conformers or strains may characterize different tauopathies, explaining pathological differences in tau diseases.
Tau strains outside the brain
A simple, non-invasive, and inexpensive test for the early detection of AD is urgently needed. It is well-established that tau levels are increased in the cerebrospinal fluid (CSF) of patients with AD(Olsson et al., 2016, Takeda S, 2016). As the central nervous system (CNS) is in direct contact with the CSF, an increase in tau oligomers in the CNS would theoretically be evident in the CSF. Furthermore, CSF is accessible via lumbar puncture in living patients. Evidence that amyloidogenic proteins may exhibit different properties in the CSF when compared to the brain was recently provided by a study that investigated seeding potential of Aβ oligomers from brain tissue and CSF. Despite a greater than 10-fold increase in levels of Aβ oligomers in the CSF compared to the brain, when injected in the brains of transgenic mice, these CSF Aβ oligomers were not capable of seeding, while those collected from the brain did seed amyloid aggregation in vivo(Fritschi et al., 2014). Therefore, differences in the conformation of oligomers in the brain versus the CSF, as well as potential heterogeneity of seeding potential of oligomer populations in the CSF may be of great importance for the development of biomarkers and diagnostic tools. Though CSF would provide a useful diagnostic tool, detecting biomarkers with a simple blood test might be preferable. Studies show that Aβ can be detected in the platelets, however high molecular weight tau that may be comprised of oligomers and other aggregates correlates better with disease symptoms(Neumann, 2011, Farías, 2012).
It is well-known that in addition to the accumulation of toxic amyloidogenic proteins in the parenchyma of the AD brain, there is also a vascular component to the disease. High levels of comorbidity between AD and vascular conditions suggest a possible common mechanism underlying vascular dysfunction and amyloidogenic protein dysregulation in AD(Yarchoan et al., 2012, Sudduth et al., 2014). Notably, animal models of AD also manifest Aβ-dependent vascular pathology(Lasagna-Reeves et al., 2014, Lin, 2014, Park et al., 2014). Vascular changes can induce hypoxia, which has been shown to facilitate the formation and deposition of Aβ and tau aggregates(Villarreal et al., 2014). The strongest correlation between vascular pathology and neurodegenerative disease is for AD, but dementia in PD and LBD has also been associated with vascular alterations(Toledo et al., 2013). Deformation of capillaries in the hippocampus and cortex is a common change in AD(Baloyannis, 2012), which could result from the over-secretion of vascular endothelial growth factor stimulated by Aβ(Dal Prà et al., 2014, Lin, 2014). Importantly, alterations in blood vessel walls may include perforations that could impair blood brain barrier (BBB) function(Scheibel, 1989). Notably, BBB impairments have been described in both neurodegenerative disease patients and mouse models(Tanifum et al., 2014).
Additionally, oligomers from amyloidogenic proteins, including Aβ, α-synuclein, and TDP43, are found in the cerebrovasculature of AD patients(Guerrero-Muñoz et al., 2014). The relationship between tau and the cerebrovasculature in tauopathies has not been thoroughly investigated, although tau has been shown to accumulate in perivascular areas affected by Aβ deposits in the AD brain(Williams et al., 2005b). Additionally, the tuft-shaped astrocytes commonly found in PSP brain frequently have vascular contact, and tau deposits are located in close proximity to blood vessels(Shibuya et al., 2011). Decreases in blood pressure in elderly patients with hypertension, which are associated with negative effects on the vasculature, and chronic cerebral hypoperfusion, which is the leading cause of vascular dementia, lead to increases in phosphorylated tau and memory deficits(Glodzik et al., 2014, Zhao et al., 2014). Moreover, tau pathology in a mouse model of tauopathy (P301L mice) is associated with brain capillary constriction prior to cell death(Jaworski et al., 2011). Collectively, the evidence suggests that vascular dysfunction is a critical component in neurodegenerative disease and that tau aggregation may play a role in this process. However, the extent to which vascular pathology varies among diseases is unknown. More research will be needed to determine whether tau aggregates may present in the cerebrovasculature and if they may differ between disorders or individuals based on conformation.
Conclusion
Evidence from the prion field and similarities between prions, other amyloidogenic proteins and tau suggest that tau likely forms different conformational strains. As is the case for prions, it is possible that the toxic entities in tauopathies include a range of strains responsible for templating and that these populations may overlap(Collinge and Clarke, 2007, Li et al., 2010). Complexity may also arise from different amyloid oligomeric seeds interacting with tau, as well as interaction with chaperones and other proteins. The characterization of tau strains may impact a variety of fields as the concept of prion-like induction and spreading of pathogenic proteins is linked to additional neurodegenerative diseases(Polymenidou and Cleveland, 2011, Munch and Bertolotti, 2012). It is important to keep in mind that the term “prion-like” refers to the self-propagation of tau and other amyloidogenic proteins across cells and tissues. It does not imply that these aggregates are infectious(Hardy and Revesz, 2012). Further multi-disciplinary study and structural characterization is needed to fully understand the most toxic tau species in disease and the impact of aggregation state and conformation on disease phenotype. The evaluation and unique detection of these structures could lead to the determination of dominant toxic strains as well as potentially neuroprotective conformers. Improved detection of tau oligomeric strains in living patients through imaging technologies such as positron emission tomography (PET) could open doors to new and more accurate diagnostic tools for neurodegenerative disease(Okamura et al.). These results could have massive impact on diagnostics as well as therapeutic approaches, thus research into this field should continue.
Acknowledgments
This work was supported by the Mitchell Center for Neurodegenerative Disease, the Sealy Center for Vaccine Development and NIH grant AG054025. We thank the members of the Kayed lab for their support and assistance.
Biographies
Dr. Julia E. Gerson is a post-doctoral fellow in Rakez Kayed’s laboratory at the University of Texas Medical Branch where she studies the toxicity of tau oligomers as they relate to neurodegenerative tauopathies.
Dr. Amrit Mudher is an Associate Professor in Neurosciences within the Centre for Biological Sciences at the University of Southampton. Her laboratory investigates the mechanisms that underpin tau-mediated dysfunction and degeneration in tauopathies such as Alzheimer’s disease and fronto-temporal dementia
Dr. Rakez Kayed is an expert on amyloid oligomers and neurodegeneration. He is an associate professor in the University of Texas Medical Branch, Department of Neurology. His laboratory studies the mechanisms of protein misfolding and aggregation using biophysical and biochemical analyses, cell and animal models and human tissue.
Footnotes
Declaration of Interest Statement
Rakez Kayed has patent applications on the compositions and methods related to tau oligomers and antibodies. No competing financial interests exist for other authors.
References
- Allen B, Ingram E, Takao M, Smith MJ, Jakes R, Virdee K, Yoshida H, Holzer M, Craxton M, Emson PC, Atzori C, Migheli A, Crowther RA, Ghetti B, Spillantini MG, Goedert M. Abundant Tau Filaments and Nonapoptotic Neurodegeneration in Transgenic Mice Expressing Human P301S Tau Protein. The Journal of Neuroscience. 2002;22:9340–9351. doi: 10.1523/JNEUROSCI.22-21-09340.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andrade-Moraes CH, Oliveira-Pinto AV, Castro-Fonseca E, Da Silva CG, Guimarães DM, Szczupak D, Parente-Bruno DR, Carvalho LRB, Polichiso L, Gomes BV, Oliveira LM, Rodriguez RD, Leite REP, Ferretti-Rebustini REL, Jacob-Filho W, Pasqualucci CA, Grinberg LT, Lent R. Cell number changes in Alzheimer’s disease relate to dementia, not to plaques and tangles. Brain. 2013;136:3738–3752. doi: 10.1093/brain/awt273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Asante EA, Smidak M, Grimshaw A, Houghton R, Tomlinson A, Jeelani A, Jakubcova T, Hamdan S, Richard-Londt A, Linehan JM, Brandner S, Alpers M, Whitfield J, Mead S, Wadsworth JDF, Collinge J. A naturally occurring variant of the human prion protein completely prevents prion disease. Nature. 2015 doi: 10.1038/nature14510. advance online publication. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballatore C, Lee VM, Trojanowski JQ. Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders. Nat Rev Neurosci. 2007;8:663–72. doi: 10.1038/nrn2194. [DOI] [PubMed] [Google Scholar]
- Baloyannis SJ, Baloyannis IS. The vascular factor in Alzheimer’s disease: A study in Golgi technique and electron microscopy. Journal of the Neurological Sciences. 2012;322:117–121. doi: 10.1016/j.jns.2012.07.010. [DOI] [PubMed] [Google Scholar]
- Barghorn S, Mandelkow E. Toward a Unified Scheme for the Aggregation of Tau into Alzheimer Paired Helical Filaments†. Biochemistry. 2002;41:14885–14896. doi: 10.1021/bi026469j. [DOI] [PubMed] [Google Scholar]
- Bartz JC, Bessen RA, Mckenzie D, Marsh RF, Aiken JM. Adaptation and selection of prion protein strain conformations following interspecies transmission of transmissible mink encephalopathy. J Virol. 2000;74:5542–5547. doi: 10.1128/jvi.74.12.5542-5547.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berger Z, Roder H, Hanna A, Carlson A, Rangachari V, Yue M, Wszolek Z, Ashe K, Knight J, Dickson D, Andorfer C, Rosenberry TL, Lewis J, Hutton M, Janus C. Accumulation of pathological tau species and memory loss in a conditional model of tauopathy. J Neurosci. 2007;27:3650–62. doi: 10.1523/JNEUROSCI.0587-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bessen RaaM, RF Identification of two biologically distinct strains of transmissible mink encephalopathy in hamsters. J Gen Virol. 1992;73:329–334. doi: 10.1099/0022-1317-73-2-329. [DOI] [PubMed] [Google Scholar]
- Bloom GS. Amyloid-β and tau: The trigger and bullet in alzheimer disease pathogenesis. JAMA Neurology. 2014;71:505–508. doi: 10.1001/jamaneurol.2013.5847. [DOI] [PubMed] [Google Scholar]
- Boluda S, Iba M, Zhang B, Raible K, Lee VY, Trojanowski J. Differential induction and spread of tau pathology in young PS19 tau transgenic mice following intracerebral injections of pathological tau from Alzheimer’s disease or corticobasal degeneration brains. Acta Neuropathologica. 2015;129:221–237. doi: 10.1007/s00401-014-1373-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braak H, Braak E. Demonstration of amyloid deposits and neurofibrillary changes in whole brain sections. Brain Pathol. 1991;1:213–6. doi: 10.1111/j.1750-3639.1991.tb00661.x. [DOI] [PubMed] [Google Scholar]
- Braak H, Braak E. Staging of alzheimer’s disease-related neurofibrillary changes. Neurobiology of Aging. 1995;16:271–278. doi: 10.1016/0197-4580(95)00021-6. [DOI] [PubMed] [Google Scholar]
- Bramblett GT, Goedert M, Jakes R, Merrick SE, Trojanowski JQ, Lee VMY. Abnormal tau phosphorylation at Ser396 in alzheimer’s disease recapitulates development and contributes to reduced microtubule binding. Neuron. 1993;10:1089–1099. doi: 10.1016/0896-6273(93)90057-x. [DOI] [PubMed] [Google Scholar]
- Brundin P, Melki R, Kopito R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat Rev Mol Cell Biol. 2010;11:301–7. doi: 10.1038/nrm2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Busciglio J, Lorenzo A, Yeh J, Yankner BA. beta-amyloid fibrils induce tau phosphorylation and loss of microtubule binding. Neuron. 1995;14:879–88. doi: 10.1016/0896-6273(95)90232-5. [DOI] [PubMed] [Google Scholar]
- Castillo-Carranza DL, Gerson JE, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Kayed R. Specific targeting of tau oligomers in htau mice prevents cognitive impairment and tau toxicity following injection with brain-derived tau oligomeric seeds. J Alzheimers Dis. 2014a;40:S97–S111. doi: 10.3233/JAD-132477. [DOI] [PubMed] [Google Scholar]
- Castillo-Carranza DL, Guerrero-Muñoz MJ, Sengupta U, Hernandez C, Barrett ADT, Dineley K, Kayed R. Tau Immunotherapy Modulates Both Pathological Tau and Upstream Amyloid Pathology in an Alzheimer’s Disease Mouse Model. The Journal of Neuroscience. 2015;35:4857–4868. doi: 10.1523/JNEUROSCI.4989-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Gerson JE, Singh G, Estes DM, Barrett AD, Dineley KT, Jackson GR, Kayed R. Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J Neurosci. 2014b;34:4260–72. doi: 10.1523/JNEUROSCI.3192-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chabrier MA, Blurton-Jones M, Agazaryan AA, Nerhus JL, Martinez-Coria H, Laferla FM. Soluble Aβ Promotes Wild-Type Tau Pathology In Vivo. The Journal of Neuroscience. 2012;32:17345–17350. doi: 10.1523/JNEUROSCI.0172-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cizas P, Budvytyte R, Morkuniene R, Moldovan R, Broccio M, Losche M, Niaura G, Valincius G, Borutaite V. Size-dependent neurotoxicity of beta-amyloid oligomers. Arch Biochem Biophys. 2010;496:84–92. doi: 10.1016/j.abb.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clavaguera F, Akatsu H, Fraser G, Crowther RA, Frank S, Hench J, Probst A, Winkler DT, Reichwald J, Staufenbiel M, Ghetti B, Goedert M, Tolnay M. Brain homogenates from human tauopathies induce tau inclusions in mouse brain. Proceedings of the National Academy of Sciences. 2013;110:9535–9540. doi: 10.1073/pnas.1301175110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clavaguera F, Bolmont T, Crowther RA, Abramowski D, Frank S, Probst A, Fraser G, Stalder AK, Beibel M, Staufenbiel M, Jucker M, Goedert M, Tolnay M. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol. 2009;11:909–13. doi: 10.1038/ncb1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clinton LK, Blurton-Jones M, Myczek K, Trojanowski JQ, Laferla FM. Synergistic Interactions between Aβ, Tau, and α-Synuclein: Acceleration of Neuropathology and Cognitive Decline. The Journal of Neuroscience. 2010;30:7281–7289. doi: 10.1523/JNEUROSCI.0490-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen ML, Kim C, Haldiman T, Elhag M, Mehndiratta P, Pichet T, Lissemore F, Shea M, Cohen Y, Chen W, Blevins J, Appleby BS, Surewicz K, Surewicz WK, Sajatovic M, Tatsuoka C, Zhang S, Mayo P, Butkiewicz M, Haines JL, Lerner AJ, Safar JG. Rapidly progressive Alzheimer’s disease features distinct structures of amyloid-β. Brain. 2015;138:1009–1022. doi: 10.1093/brain/awv006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collinge J, Clarke AR. A general model of prion strains and their pathogenicity. Science. 2007;318:930–6. doi: 10.1126/science.1138718. [DOI] [PubMed] [Google Scholar]
- Congdon EE, Kim S, Bonchak J, Songrug T, Matzavinos A, Kuret J. Nucleation-dependent tau filament formation: the importance of dimerization and an estimation of elementary rate constants. J Biol Chem. 2008;283:13806–16. doi: 10.1074/jbc.M800247200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cook C, Kang SS, Carlomagno Y, Lin WL, Yue M, Kurti A, Shinohara M, Jansen-West K, Perkerson E, Castanedes-Casey M, Rousseau L, Phillips V, Bu G, Dickson DW, Petrucelli L, Fryer JD. Tau deposition drives neuropathological, inflammatory and behavioral abnormalities independently of neuronal loss in a novel mouse model. Human Molecular Genetics. 2015;24:6198–6212. doi: 10.1093/hmg/ddv336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cowan CM, Mudher A. Are tau aggregates toxic or protective in tauopathies? Frontiers in Neurology. 2013:4. doi: 10.3389/fneur.2013.00114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cowan CM, Quraishe S, Hands S, Sealey M, Mahajan S, Allan DW, Mudher A. Rescue from tau-induced neuronal dysfunction produces insoluble tau oligomers. Scientific Reports. 2015;5:17191. doi: 10.1038/srep17191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cowan CM, Quraishe S, Mudher A. What is the pathological significance of tau oligomers? Biochem Soc Trans. 2012;40:693–7. doi: 10.1042/BST20120135. [DOI] [PubMed] [Google Scholar]
- Dal Prà I, Armato U, Chioffi F, Pacchiana R, Whitfield J, Chakravarthy B, Gui L, Chiarini A. The Aβ Peptides-Activated Calcium-Sensing Receptor Stimulates the Production and Secretion of Vascular Endothelial Growth Factor-A by Normoxic Adult Human Cortical Astrocytes. NeuroMolecular Medicine. 2014:1–13. doi: 10.1007/s12017-014-8315-9. [DOI] [PubMed] [Google Scholar]
- Danzer KM, Krebs SK, Wolff M, Birk G, Hengerer B. Seeding induced by alpha-synuclein oligomers provides evidence for spreading of alpha-synuclein pathology. J Neurochem. 2009;111:192–203. doi: 10.1111/j.1471-4159.2009.06324.x. [DOI] [PubMed] [Google Scholar]
- De Strooper B. Proteases and Proteolysis in Alzheimer Disease: A Multifactorial View on the Disease Process. 2010 doi: 10.1152/physrev.00023.2009. [DOI] [PubMed] [Google Scholar]
- Delacourte A, Sergeant N, Wattez A, Maurage CA, Lebert F, Pasquier F, David JP. Tau aggregation in the hippocampal formation: an ageing or a pathological process? Exp Gerontol. 2002;37:1291–6. doi: 10.1016/s0531-5565(02)00141-9. [DOI] [PubMed] [Google Scholar]
- Díaz-Hernández M, Gómez-Ramos A, Rubio A, Gómez-Villafuertes R, Naranjo JR, Miras-Portugal MT, Avila J. Tissue-nonspecific Alkaline Phosphatase Promotes the Neurotoxicity Effect of Extracellular Tau. Journal of Biological Chemistry. 2010;285:32539–32548. doi: 10.1074/jbc.M110.145003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dickey C, Yue M, Lin W, Dickson D, Dunmore J, Lee W, Zehr C, West G, Cao S, Clark A, Caldwell G, Caldwell K, Eckman C, Patterson C, Hutton M, Petrucelli L. Deletion of the ubiquitin ligase CHIP leads to the accumulation, but not the aggregation, of both endogenous phospho- and caspase-3-cleaved tau species. J Neurosci. 2006;26:6985– 6996. doi: 10.1523/JNEUROSCI.0746-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dinkel PD, Siddiqua A, Huynh H, Shah M, Margittai M. Variations in filament conformation dictate seeding barrier between three- and four-repeat tau. Biochemistry. 2011;50:4330–6. doi: 10.1021/bi2004685. [DOI] [PubMed] [Google Scholar]
- Farías G, Pérez P, Slachevsky A, Maccioni RB. Platelet Tau Pattern Correlates with Cognitive Status in Alzheimer’s Disease. J Alzheimers Dis. 2012;31:65–69. doi: 10.3233/JAD-2012-120304. [DOI] [PubMed] [Google Scholar]
- Ferrari A, Hoerndli F, Baechi T, Nitsch RM, Gotz J. beta-Amyloid induces paired helical filament-like tau filaments in tissue culture. J Biol Chem. 2003;278:40162–8. doi: 10.1074/jbc.M308243200. [DOI] [PubMed] [Google Scholar]
- Fritschi SK, Langer F, Kaeser SA, Maia LF, Portelius E, Pinotsi D, Kaminski CF, Winkler DT, Maetzler W, Keyvani K, Spitzer P, Wiltfang J, Kaminski Schierle GS, Zetterberg H, Staufenbiel M, Jucker M. Highly potent soluble amyloid-β seeds in human Alzheimer brain but not cerebrospinal fluid. Brain. 2014 doi: 10.1093/brain/awu255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11:155–9. doi: 10.1038/nrn2786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Furukawa Y, Kaneko K, Nukina N. Tau Protein Assembles into Isoform- and Disulfide-dependent Polymorphic Fibrils with Distinct Structural Properties. Journal of Biological Chemistry. 2011;286:27236–27246. doi: 10.1074/jbc.M111.248963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gendron TF, Petrucelli L. The role of tau in neurodegeneration. Mol Neurodegener. 2009;4:13. doi: 10.1186/1750-1326-4-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerson JE, Castillo-Carranza DL, Kayed R. Advances in Therapeutics for Neurodegenerative Tauopathies: Moving toward the Specific Targeting of the Most Toxic Tau Species. ACS Chemical Neuroscience. 2014;5:752–769. doi: 10.1021/cn500143n. [DOI] [PubMed] [Google Scholar]
- Giasson BI, Forman MS, Higuchi M, Golbe LI, Graves CL, Kotzbauer PT, Trojanowski JQ, Lee VM. Initiation and synergistic fibrillization of tau and alpha-synuclein. Science. 2003;300:636–40. doi: 10.1126/science.1082324. [DOI] [PubMed] [Google Scholar]
- Gispert S, Del Turco D, Garrett L, Chen A, Bernard DJ, Hamm-Clement J, Korf HW, Deller T, Braak H, Auburger G, Nussbaum RL. Transgenic mice expressing mutant A53T human alpha-synuclein show neuronal dysfunction in the absence of aggregate formation. Mol Cell Neurosci. 2003;24:419–29. doi: 10.1016/s1044-7431(03)00198-2. [DOI] [PubMed] [Google Scholar]
- Glabe CG. Structural classification of toxic amyloid oligomers. J Biol Chem. 2008;283:29639–43. doi: 10.1074/jbc.R800016200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glabe CG, Kayed R. Common structure and toxic function of amyloid oligomers implies a common mechanism of pathogenesis. Neurology. 2006;66:S74–8. doi: 10.1212/01.wnl.0000192103.24796.42. [DOI] [PubMed] [Google Scholar]
- Glodzik L, Rusinek H, Pirraglia E, Mchugh P, Tsui W, Williams S, Cummings M, Li Y, Rich K, Randall C, Mosconi L, Osorio R, Murray J, Zetterberg H, Blennow K, De Leon M. Blood pressure decrease correlates with tau pathology and memory decline in hypertensive elderly. Neurobiology of Aging. 2014;35:64–71. doi: 10.1016/j.neurobiolaging.2013.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Godyń J, Jończyk J, Panek D, Malawska B. Therapeutic strategies for Alzheimer’s disease in clinical trials. Pharmacological Reports. 2016;68:127–138. doi: 10.1016/j.pharep.2015.07.006. [DOI] [PubMed] [Google Scholar]
- Goedert M, Jakes R. Mutations causing neurodegenerative tauopathies. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2005;1739:240–250. doi: 10.1016/j.bbadis.2004.08.007. [DOI] [PubMed] [Google Scholar]
- Goedert M, Jakes R, Crowther RA. Effects of frontotemporal dementia FTDP-17 mutations on heparin-induced assembly of tau filaments. FEBS Letters. 1999;450:306–311. doi: 10.1016/s0014-5793(99)00508-6. [DOI] [PubMed] [Google Scholar]
- Goedert M, Jakes R, Spillantini MG, Hasegawa M, Smith MJ, Crowther RA. Assembly of microtubule-associated protein tau into Alzheimer-like filaments induced by sulphated glycosaminoglycans. Nature. 1996;383:550–553. doi: 10.1038/383550a0. [DOI] [PubMed] [Google Scholar]
- Götz J, Chen F, Barmettler R, Nitsch RM. Tau Filament Formation in Transgenic Mice Expressing P301L Tau. Journal of Biological Chemistry. 2001;276:529–534. doi: 10.1074/jbc.M006531200. [DOI] [PubMed] [Google Scholar]
- Gotz J, Chen F, Van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science. 2001;293:1491–5. doi: 10.1126/science.1062097. [DOI] [PubMed] [Google Scholar]
- Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proceedings of the National Academy of Sciences. 1986;83:4913–4917. doi: 10.1073/pnas.83.13.4913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guerrero-Muñoz MJ, Castillo-Carranza DL, Krishnamurthy S, Paulucci-Holthauzen AA, Sengupta U, Lasagna-Reeves CA, Ahmad Y, Jackson GR, Kayed R. Amyloid-β oligomers as a template for secondary amyloidosis in Alzheimer’s disease. Neurobiology of Disease. 2014;71:14–23. doi: 10.1016/j.nbd.2014.08.008. [DOI] [PubMed] [Google Scholar]
- Guo Jl, Covell Dj, Daniels Jp, Iba M, Stieber A, Zhang B, Riddle Dm, Kwong Lk, Xu Y, Trojanowski Jq, Lee VmY. Distinct α-Synuclein Strains Differentially Promote Tau Inclusions in Neurons. Cell. 2013;154:103–117. doi: 10.1016/j.cell.2013.05.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Habicht G, Haupt C, Friedrich RP, Hortschansky P, Sachse C, Meinhardt J, Wieligmann K, Gellermann GP, Brodhun M, Götz J, Halbhuber KJ, Röcken C, Horn U, Fändrich M. Directed selection of a conformational antibody domain that prevents mature amyloid fibril formation by stabilizing Aβ protofibrils. Proceedings of the National Academy of Sciences. 2007;104:19232–19237. doi: 10.1073/pnas.0703793104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hafner JH, Cheung CL, Woolley AT, Lieber CM. Structural and functional imaging with carbon nanotube AFM probes. Progress in Biophysics and Molecular Biology. 2001;77:73–110. doi: 10.1016/s0079-6107(01)00011-6. [DOI] [PubMed] [Google Scholar]
- Hardy J, Revesz T. The spread of neurodegenerative disease. N Engl J Med. 2012;366:2126–8. doi: 10.1056/NEJMcibr1202401. [DOI] [PubMed] [Google Scholar]
- Hardy JDJ. The Amyloid Hypothesis of Alzheimer’s Disease: Progress and Problems on the Road to Therapeutics. Science. 2002;297:353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
- Haroutunian V, Davies P, Vianna C, Buxbaum JD, Purohit DP. Tau protein abnormalities associated with the progression of alzheimer disease type dementia. Neurobiol Aging. 2007;28:1–7. doi: 10.1016/j.neurobiolaging.2005.11.001. [DOI] [PubMed] [Google Scholar]
- Harper JD, Wong SS, Lieber CM, Lansbury PT., Jr Observation of metastable Aβ amyloid protofibrils by atomic force microscopy. Chemistry & Biology. 1997;4:119–125. doi: 10.1016/s1074-5521(97)90255-6. [DOI] [PubMed] [Google Scholar]
- Heilbronner GEY, Langer F, Kaeser Sa, Novotny R, Nagarathinam a, Aslund a, Hammarström P, Nilsson Kp, Jucker M. Seeded strain-like transmission of β-amyloid morphotypes in APP transgenic mice. EMBO Rep. 2013;14:1017–1022. doi: 10.1038/embor.2013.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ho L, Xiang Z, Mukherjee P, Zhang W, De Jesus N, Mirjany M, Yemul S, Pasinetti GM. Gene expression profiling of the tau mutant (P301L) transgenic mouse brain. Neuroscience Letters. 2001;310:1–4. doi: 10.1016/s0304-3940(01)02044-4. [DOI] [PubMed] [Google Scholar]
- Hyman Bradley t. Tau Propagation, Different Tau Phenotypes, and Prion-like Properties of Tau. Neuron. 2014;82:1189–1190. doi: 10.1016/j.neuron.2014.06.004. [DOI] [PubMed] [Google Scholar]
- Iseki E, Takayama N, Marui W, Ueda K, Kosaka K. Relationship in the formation process between neurofibrillary tangles and Lewy bodies in the hippocampus of dementia with Lewy bodies brains. J Neurol Sci. 2002;195:85–91. doi: 10.1016/s0022-510x(01)00689-x. [DOI] [PubMed] [Google Scholar]
- Jarrett JT, Berger EP, Lansbury PT., Jr The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer’s disease. Biochemistry. 1993;32:4693–7. doi: 10.1021/bi00069a001. [DOI] [PubMed] [Google Scholar]
- Jaworski T, Lechat B, Demedts D, Gielis L, Devijver H, Borghgraef P, Duimel H, Verheyen F, Kügler S, Van Leuven F. Dendritic Degeneration, Neurovascular Defects, and Inflammation Precede Neuronal Loss in a Mouse Model for Tau-Mediated Neurodegeneration. The American Journal of Pathology. 2011;179:2001–2015. doi: 10.1016/j.ajpath.2011.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jensen PH, Hager H, Nielsen MS, Hojrup P, Gliemann J, Jakes R. alpha-synuclein binds to Tau and stimulates the protein kinase A-catalyzed tau phosphorylation of serine residues 262 and 356. J Biol Chem. 1999;274:25481–9. doi: 10.1074/jbc.274.36.25481. [DOI] [PubMed] [Google Scholar]
- Kanmert D, Cantlon A, Muratore CR, Jin M, O’malley TT, Lee G, Young-Pearse TL, Selkoe DJ, Walsh DM. C-Terminally Truncated Forms of Tau, But Not Full-Length Tau or Its C-Terminal Fragments, Are Released from Neurons Independently of Cell Death. The Journal of Neuroscience. 2015;35:10851–10865. doi: 10.1523/JNEUROSCI.0387-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kayed R, Canto I, Breydo L, Rasool S, Lukacsovich T, Wu J, Albay R, 3rd, Pensalfini A, Yeung S, Head E, Marsh JL, Glabe C. Conformation dependent monoclonal antibodies distinguish different replicating strains or conformers of prefibrillar Abeta oligomers. Mol Neurodegener. 2010;5:57. doi: 10.1186/1750-1326-5-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kayed R, Glabe CG. Conformation-dependent anti-amyloid oligomer antibodies. Methods Enzymol. 2006;413:326–44. doi: 10.1016/S0076-6879(06)13017-7. [DOI] [PubMed] [Google Scholar]
- Kayed R, Head E, Sarsoza F, Saing T, Cotman CW, Necula M, Margol L, Wu J, Breydo L, Thompson JL, Rasool S, Gurlo T, Butler P, Glabe CG. Fibril specific, conformation dependent antibodies recognize a generic epitope common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar oligomers. Mol Neurodegener. 2007;2:18. doi: 10.1186/1750-1326-2-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kayed R, Head E, Thompson JL, Mcintire TM, Milton SC, Cotman CW, Glabe CG. Common Structure of Soluble Amyloid Oligomers Implies Common Mechanism of Pathogenesis. Science. 2003;300:486–489. doi: 10.1126/science.1079469. [DOI] [PubMed] [Google Scholar]
- Kayed R, Lasagna-Reeves CA. Molecular mechanisms of amyloid oligomers toxicity. J Alzheimers Dis. 2013;33(Suppl 1):S67–78. doi: 10.3233/JAD-2012-129001. [DOI] [PubMed] [Google Scholar]
- Kelly JW. Mechanisms of amyloidogenesis. Nat Struct Biol. 2000;7:824–6. doi: 10.1038/82815. [DOI] [PubMed] [Google Scholar]
- Kim Y, Choi H, Lee W, Park H, Kam TI, Hong SH, Nah J, Jung S, Shin B, Lee H, Choi TY, Choo H, Kim KK, Choi SY, Kayed R, Jung Y-K. Caspase-cleaved tau exhibits rapid memory impairment associated with tau oligomers in a transgenic mouse model. Neurobiology of Disease. 2016;87:19–28. doi: 10.1016/j.nbd.2015.12.006. [DOI] [PubMed] [Google Scholar]
- Klein WL, Krafft GA, Finch CE. Targeting small Abeta oligomers: the solution to an Alzheimer’s disease conundrum? Trends Neurosci. 2001;24:219–24. doi: 10.1016/s0166-2236(00)01749-5. [DOI] [PubMed] [Google Scholar]
- Kopeikina KJ, Carlson GA, Pitstick R, Ludvigson AE, Peters A, Luebke JI, Koffie RM, Frosch MP, Hyman BT, Spires-Jones TL. Tau accumulation causes mitochondrial distribution deficits in neurons in a mouse model of tauopathy and in human Alzheimer’s disease brain. Am J Pathol. 2011;179:2071–82. doi: 10.1016/j.ajpath.2011.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kril JJ, Patel S, Harding AJ, Halliday GM. Neuron loss from the hippocampus of Alzheimer’s disease exceeds extracellular neurofibrillary tangle formation. Acta Neuropathol. 2002;103:370–6. doi: 10.1007/s00401-001-0477-5. [DOI] [PubMed] [Google Scholar]
- Ksiezak-Reding H, Morgan K, Mattiace LA, Davies P, Liu WK, Yen SH, Weidenheim K, Dickson DW. Ultrastructure and biochemical composition of paired helical filaments in corticobasal degeneration. Am J Pathol. 1994;145:1496–1508. [PMC free article] [PubMed] [Google Scholar]
- Langer F, Eisele YS, Fritschi SK, Staufenbiel M, Walker LC, Jucker M. Soluble Abeta seeds are potent inducers of cerebral beta-amyloid deposition. J Neurosci. 2011;31:14488–95. doi: 10.1523/JNEUROSCI.3088-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lasagna-Reeves C, Sengupta U, Castillo-Carranza D, Gerson J, Guerrero-Munoz M, Troncoso J, Jackson G, Kayed R. The formation of tau pore-like structures is prevalent and cell specific: possible implications for the disease phenotypes. Acta Neuropathologica Communications. 2014;2:56. doi: 10.1186/2051-5960-2-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lasagna-Reeves CA, Castillo-Carranza DL, Guerrero-Muoz MJ, Jackson GR, Kayed R. Preparation and characterization of neurotoxic tau oligomers. Biochemistry. 2010;49:10039–41. doi: 10.1021/bi1016233. [DOI] [PubMed] [Google Scholar]
- Lasagna-Reeves CA, Castillo-Carranza DL, Jackson GR, Kayed R. Tau oligomers as potential targets for immunotherapy for Alzheimer’s disease and tauopathies. Curr Alzheimer Res. 2011a;8:659–65. doi: 10.2174/156720511796717177. [DOI] [PubMed] [Google Scholar]
- Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Clos AL, Jackson GR, Kayed R. Tau Oligomers Impair Memory and Induce Synaptic and Mitochondrial Dysfunction in Wild-type Mice. Mol Neurodegener. 2011b;6:39. doi: 10.1186/1750-1326-6-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Kiritoshi T, Neugebauer V, Jackson GR, Kayed R. Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Sci Rep. 2012a;2:700. doi: 10.1038/srep00700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Sarmiento J, Troncoso J, Jackson GR, Kayed R. Identification of oligomers at early stages of tau aggregation in Alzheimer’s disease. FASEB J. 2012b doi: 10.1096/fj.11-199851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lasagna-Reeves CA, Glabe CG, Kayed R. Amyloid-β Annular Protofibrils Evade Fibrillar Fate in Alzheimer Disease Brain. Journal of Biological Chemistry. 2011c;286:22122–22130. doi: 10.1074/jbc.M111.236257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J, Culyba EK, Powers ET, Kelly JW. Amyloid-beta forms fibrils by nucleated conformational conversion of oligomers. Nat Chem Biol. 2011;7:602–9. doi: 10.1038/nchembio.624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee VMY, Balin BJ, Otvos L, Jr, Trojanowski JQ. A68: A Major Subunit of Paired Helical Filaments and Derivatized Forms of Normal Tau. Science. 1991;251:675–678. doi: 10.1126/science.1899488. [DOI] [PubMed] [Google Scholar]
- Legname G, Nguyen HOB, Baskakov IV, Cohen FE, Dearmond SJ, Prusiner SB. Strain-specified characteristics of mouse synthetic prions. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:2168–2173. doi: 10.1073/pnas.0409079102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, Gallagher M, Ashe KH. A specific amyloid-beta protein assembly in the brain impairs memory. Nature. 2006;440:352–7. doi: 10.1038/nature04533. [DOI] [PubMed] [Google Scholar]
- Lewis JME, Rockwood J, Melrose H, Nacharaju P, Van Slegtenhorst M, Gwinn-Hardy K, Paul Murphy M, Baker M, Yu X, Duff K, Hardy J, Corral a, Lin Wl, Yen Sh, Dickson Dw, Davies P, Hutton M. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet. 2000;25:402–405. doi: 10.1038/78078. [DOI] [PubMed] [Google Scholar]
- Li J, Browning S, Mahal SP, Oelschlegel AM, Weissmann C. Darwinian evolution of prions in cell culture. Science. 2010;327:869–72. doi: 10.1126/science.1183218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin AJ, Liu G, Castello NA, Yeh JJ, Rahimian R, Lee G, Tsay V, Durkin AJ, Choi B, Laferla FM, Chen Z, Green KN, Tromberg BJ. Optical imaging in an Alzheimer’s mouse model reveals amyloid-β-dependent vascular impairment. Neurophotonics. 2014;1:011005. doi: 10.1117/1.NPh.1.1.011005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu P, Reed Miranda n, Kotilinek Linda a, Grant Marianne kO, Forster Colleen l, Qiang W, Shapiro Samantha l, Reichl John h, Chiang Angie cA, Jankowsky Joanna l, Wilmot Carrie m, Cleary James p, Zahs Kathleen r, Ashe, Karen h. Quaternary Structure Defines a Large Class of Amyloid-β Oligomers Neutralized by Sequestration. Cell Reports. 2015;11:1760–1771. doi: 10.1016/j.celrep.2015.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu WK, Dickson DW, Yen SH. Heterogeneity of tau proteins in Alzheimer’s disease. Evidence for increased expression of an isoform and preferential distribution of a phosphorylated isoform in neurites. Am J Pathol. 1993;142:387–394. [PMC free article] [PubMed] [Google Scholar]
- Lu Jx, QW, Yau Wm, Schwieters Cd, Meredith Sc, Tycko R. Molecular structure of β-amyloid fibrils in Alzheimer’s disease brain tissue. Cell. 2013;154:1257–1268. doi: 10.1016/j.cell.2013.08.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maeda S, Sahara N, Saito Y, Murayama M, Yoshiike Y, Kim H, Miyasaka T, Murayama S, Ikai A, Takashima A. Granular tau oligomers as intermediates of tau filaments. Biochemistry. 2007;46:3856–61. doi: 10.1021/bi061359o. [DOI] [PubMed] [Google Scholar]
- Maeda S, Sahara N, Saito Y, Murayama S, Ikai A, Takashima A. Increased levels of granular tau oligomers: an early sign of brain aging and Alzheimer’s disease. Neurosci Res. 2006;54:197–201. doi: 10.1016/j.neures.2005.11.009. [DOI] [PubMed] [Google Scholar]
- Margittai M, Langen R. Template-assisted filament growth by parallel stacking of tau. Proc Natl Acad Sci U S A. 2004;101:10278–83. doi: 10.1073/pnas.0401911101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martins IC, Kuperstein I, Wilkinson H, Maes E, Vanbrabant M, Jonckheere W, Van Gelder P, Hartmann D, D’hooge R, De Strooper B, Schymkowitz J, Rousseau F. Lipids revert inert Aβ amyloid fibrils to neurotoxic protofibrils that affect learning in mice. 2008 doi: 10.1038/sj.emboj.7601953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsumoto SE, Motoi Y, Ishiguro K, Tabira T, Kametani F, Hasegawa M, Hattori N. The twenty-four KDa C-terminal tau fragment increases with aging in tauopathy mice: implications of prion-like properties. Human Molecular Genetics. 2015;24:6403–6416. doi: 10.1093/hmg/ddv351. [DOI] [PubMed] [Google Scholar]
- Meier S, Bell M, Lyons DN, Ingram A, Chen J, Gensel JC, Zhu H, Nelson PT, Abisambra JF. Identification of Novel Tau Interactions with Endoplasmic Reticulum Proteins in Alzheimer’s Disease Brain. J Alzheimers Dis. 2015;48:687–702. doi: 10.3233/JAD-150298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer V, Dinkel PD, Luo Y, Yu X, Wei G, Zheng J, Eaton GR, Ma B, Nussinov R, Eaton SS, Margittai M. Single mutations in tau modulate the populations of fibril conformers through seed selection. Angew Chem Int Ed Engl. 2014;53:1590–3. doi: 10.1002/anie.201308473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller BE. Comparison of A68 levels in Alzheimer Diseased and non-Alzheimer’s diseased brain by two ALZ50 based methods. Life Sciences. 1999;65:2215–2222. doi: 10.1016/s0024-3205(99)00486-5. [DOI] [PubMed] [Google Scholar]
- Mina EW, Lasagna-Reeves C, Glabe CG, Kayed R. Poloxamer 188 copolymer membrane sealant rescues toxicity of amyloid oligomers in vitro. J Mol Biol. 2009;391:577–85. doi: 10.1016/j.jmb.2009.06.024. [DOI] [PubMed] [Google Scholar]
- Morales R, Abid K, Soto C. The prion strain phenomenon: Molecular basis and unprecedented features. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2007;1772:681–691. doi: 10.1016/j.bbadis.2006.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Munch C, Bertolotti A. Propagation of the prion phenomenon: beyond the seeding principle. J Mol Biol. 2012;421:491–8. doi: 10.1016/j.jmb.2011.12.061. [DOI] [PubMed] [Google Scholar]
- Munoz-Garcia D. Classic and generalized variants of Pick’s disease: a clinicopathological, ultrastructural, and immunocytochemical comparative study. Annals of neurology. 1984;16:467–80. doi: 10.1002/ana.410160408. [DOI] [PubMed] [Google Scholar]
- Necula M, Kayed R, Milton S, Glabe CG. Small molecule inhibitors of aggregation indicate that amyloid beta oligomerization and fibrillization pathways are independent and distinct. J Biol Chem. 2007;282:10311–24. doi: 10.1074/jbc.M608207200. [DOI] [PubMed] [Google Scholar]
- Neumann K, Farías G, Slachevsky A, Perez P, Maccioni RB. Human Platelets Tau: A Potential Peripheral Marker for Alzheimer’s Disease. J Alzheimers Dis. 2011;25:103–109. doi: 10.3233/JAD-2011-101641. [DOI] [PubMed] [Google Scholar]
- O’nuallain B, Williams AD, Westermark P, Wetzel R. Seeding specificity in amyloid growth induced by heterologous fibrils. J Biol Chem. 2004;279:17490–9. doi: 10.1074/jbc.M311300200. [DOI] [PubMed] [Google Scholar]
- Oddo S, Vasilevko V, Caccamo A, Kitazawa M, Cribbs DH, Laferla FM. Reduction of Soluble Aβ and Tau, but Not Soluble Aβ Alone, Ameliorates Cognitive Decline in Transgenic Mice with Plaques and Tangles. Journal of Biological Chemistry. 2006;281:39413–39423. doi: 10.1074/jbc.M608485200. [DOI] [PubMed] [Google Scholar]
- Okamura N, Harada R, Furukawa K, Furumoto S, Tago T, Yanai K, Arai H, Kudo Y. Advances in the development of tau PET radiotracers and their clinical applications. Ageing Research Reviews. doi: 10.1016/j.arr.2015.12.010. [DOI] [PubMed] [Google Scholar]
- Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, Hölttä M, Rosén C, Olsson C, Strobel G, Wu E, Dakin K, Petzold M, Blennow K, Zetterberg H. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. The Lancet Neurology. 2016;15:673–684. doi: 10.1016/S1474-4422(16)00070-3. [DOI] [PubMed] [Google Scholar]
- Papanikolopoulou K, Skoulakis EMC. Temporally distinct phosphorylations differentiate Tau-dependent learning deficits and premature mortality in Drosophila. Human Molecular Genetics. 2015;24:2065–2077. doi: 10.1093/hmg/ddu726. [DOI] [PubMed] [Google Scholar]
- Park L, Koizumi K, El Jamal S, Zhou P, Previti M, Van Nostrand WE, Carlson G, Iadecola C. Age-Dependent Neurovascular Dysfunction and Damage in a Mouse Model of Cerebral Amyloid Angiopathy. Stroke. 2014;45:1815–1821. doi: 10.1161/STROKEAHA.114.005179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peelaerts W, Bousset L, Van Der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, Van Den Haute C, Melki R, Baekelandt V. [agr]-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015 doi: 10.1038/nature14547. advance online publication. [DOI] [PubMed] [Google Scholar]
- Pennanen L, Gotz J. Different tau epitopes define Abeta42-mediated tau insolubility. Biochem Biophys Res Commun. 2005;337:1097–101. doi: 10.1016/j.bbrc.2005.09.168. [DOI] [PubMed] [Google Scholar]
- Petrucelli L, Dickson D, Kehoe K, Taylor J, Snyder H, Grover A, De Lucia M, Mcgowan E, Lewis J, Prihar G, Kim J, Dillmann WH, Browne SE, Hall A, Voellmy R, Tsuboi Y, Dawson TM, Wolozin B, Hardy J, Hutton M. CHIP and Hsp70 regulate tau ubiquitination, degradation and aggregation. Human Molecular Genetics. 2004;13:703–714. doi: 10.1093/hmg/ddh083. [DOI] [PubMed] [Google Scholar]
- Polymenidou M, Cleveland DW. The seeds of neurodegeneration: prion-like spreading in ALS. Cell. 2011;147:498–508. doi: 10.1016/j.cell.2011.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prusiner SB, Woerman AL, Mordes DA, Watts JC, Rampersaud R, Berry DB, Patel S, Oehler A, Lowe JK, Kravitz SN, Geschwind DH, Glidden DV, Halliday GM, Middleton LT, Gentleman SM, Grinberg LT, Giles K. Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proceedings of the National Academy of Sciences. 2015;112:E5308–E5317. doi: 10.1073/pnas.1514475112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reifert J, Hartung-Cranston D, Feinstein SC. Amyloid β-Mediated Cell Death of Cultured Hippocampal Neurons Reveals Extensive Tau Fragmentation without Increased Full-length Tau Phosphorylation. Journal of Biological Chemistry. 2011;286:20797–20811. doi: 10.1074/jbc.M111.234674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rijal Upadhaya A, Capetillo-Zarate E, Kosterin I, Abramowski D, Kumar S, Yamaguchi H, Walter J, Fändrich M, Staufenbiel M, Thal DR. Dispersible amyloid β-protein oligomers, protofibrils, and fibrils represent diffusible but not soluble aggregates: their role in neurodegeneration in amyloid precursor protein (APP) transgenic mice. Neurobiology of Aging. 2012;33:2641–2660. doi: 10.1016/j.neurobiolaging.2011.12.032. [DOI] [PubMed] [Google Scholar]
- Roberts RF, Wade-Martins R, Alegre-Abarrategui J. Direct visualization of alpha-synuclein oligomers reveals previously undetected pathology in Parkinson’s disease brain. Brain. 2015;138:1642–1657. doi: 10.1093/brain/awv040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahara N, Deture M, Ren Y, Ebrahim AS, Kang D, Knight J, Volbracht C, Pedersen JT, Dickson DW, Yen SH, Lewis J. Characteristics of TBS-extractable hyperphosphorylated Tau species: aggregation intermediates in rTg4510 mouse brain. J Alzheimers Dis. 2013;33:249–63. doi: 10.3233/JAD-2012-121093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanders D, Kaufman Sk, Devos Sl, Sharma Am, Mirbaha H, Li A, Barker Sj, Foley A, Thorpe, Serpell Lc, Miller Tm, Grinberg Lt, Seeley Ww, Diamond Mi. Distinct Tau Prion Strains Propagate in Cells and Mice and Define Different Tauopathies. Neuron. 2014 doi: 10.1016/j.neuron.2014.04.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheibel AB, Duong T, Jacobs R. Alzheimer’s disease as a capillary dementia. Annals of Medicine. 1989;21:103–107. doi: 10.3109/07853898909149194. [DOI] [PubMed] [Google Scholar]
- Schoch G, Seeger H, Bogousslavsky J, Tolnay M, Janzer RC, Aguzzi A, Glatzel M. Analysis of prion strains by PrPSc profiling in sporadic Creutzfeldt-Jakob disease. PLoS Med. 2006;3:e14. doi: 10.1371/journal.pmed.0030014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sengupta U, Guerrero-Muñoz MJ, Castillo-Carranza DL, Lasagna-Reeves CA, Gerson JE, Paulucci-Holthauzen AA, Krishnamurthy S, Farhed M, Jackson GR, Kayed R. Pathological Interface between Oligomeric Alpha-Synuclein and Tau in Synucleinopathies. Biological Psychiatry. 2015 doi: 10.1016/j.biopsych.2014.12.019. [DOI] [PubMed] [Google Scholar]
- Shibuya K, Yagishita S, Nakamura A, Uchihara T. Perivascular orientation of astrocytic plaques and tuft-shaped astrocytes. Brain Research. 2011;1404:50–54. doi: 10.1016/j.brainres.2011.06.014. [DOI] [PubMed] [Google Scholar]
- Shin RW, Bramblett GT, Lee VM, Trojanowski JQ. Alzheimer disease A68 proteins injected into rat brain induce codeposits of beta-amyloid, ubiquitin, and alpha 1-antichymotrypsin. Proceedings of the National Academy of Sciences. 1993;90:6825–6828. doi: 10.1073/pnas.90.14.6825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siddiqua A, Margittai M. Three- and four-repeat Tau coassemble into heterogeneous filaments: an implication for Alzheimer disease. J Biol Chem. 2010;285:37920–6. doi: 10.1074/jbc.M110.185728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Šimić G, Babić Leko M, Wray S, Harrington C, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, De Silva R, Di Giovanni G, Wischik C, Hof P. Tau Protein Hyperphosphorylation and Aggregation in Alzheimer’s Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules. 2016;6:6. doi: 10.3390/biom6010006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simon-Sanchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, Paisan-Ruiz C, Lichtner P, Scholz SW, Hernandez DG, Kruger R, Federoff M, Klein C, Goate A, Perlmutter J, Bonin M, Nalls MA, Illig T, Gieger C, Houlden H, Steffens M, Okun MS, Racette BA, Cookson MR, Foote KD, Fernandez HH, Traynor BJ, Schreiber S, Arepalli S, Zonozi R, Gwinn K, Van Der Brug M, Lopez G, Chanock SJ, Schatzkin A, Park Y, Hollenbeck A, Gao J, Huang X, Wood NW, Lorenz D, Deuschl G, Chen H, Riess O, Hardy JA, Singleton AB, Gasser T. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet. 2009;41:1308–12. doi: 10.1038/ng.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sneideris T, Darguzis D, Botyriute A, Grigaliunas M, Winter R, Smirnovas V. pH-Driven Polymorphism of Insulin Amyloid-Like Fibrils. PLoS ONE. 2015;10:e0136602. doi: 10.1371/journal.pone.0136602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spires TL, Orne JD, Santacruz K, Pitstick R, Carlson GA, Ashe KH, Hyman BT. Region-specific dissociation of neuronal loss and neurofibrillary pathology in a mouse model of tauopathy. Am J Pathol. 2006;168:1598–607. doi: 10.2353/ajpath.2006.050840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sporn ZA, Hines JK. Hsp40 function in yeast prion propagation: Amyloid diversity necessitates chaperone functional complexity. Prion. 2015;9:80–89. doi: 10.1080/19336896.2015.1020268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sudduth T, Weekman E, Brothers H, Braun K, Wilcock D. beta-amyloid deposition is shifted to the vasculature and memory impairment is exacerbated when hyperhomocysteinemia is induced in APP/PS1 transgenic mice. Alzheimer’s Research & Therapy. 2014;6:32. doi: 10.1186/alzrt262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sultan A, Nesslany F, Violet M, Bégard S, Loyens A, Talahari S, Mansuroglu Z, Marzin D, Sergeant N, Humez S, Colin M, Bonnefoy E, Buée L, Galas M-C. Nuclear Tau, a Key Player in Neuronal DNA Protection. Journal of Biological Chemistry. 2011;286:4566–4575. doi: 10.1074/jbc.M110.199976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takeda SCC, Devos Sl, Nobuhara Ck, Wegmann S, Roe Ad, Costantino I, Fan Z, Nicholls Sb, Sherman Ae, Trisini Lipsanopoulos at, Scherzer Cr, Carlson Ga, Pitstick R, Peskind Er, Raskind Ma, Li G, Montine Tj, Frosch Mp, Hyman Bt. Seed-competent HMW tau species accumulates in the cerebrospinal fluid of Alzheimer’s disease mouse model and human patients. Ann Neurol. 2016 doi: 10.1002/ana.24716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka M, Collins SR, Toyama BH, Weissman JS. The physical basis of how prion conformations determine strain phenotypes. Nature. 2006;442:585–9. doi: 10.1038/nature04922. [DOI] [PubMed] [Google Scholar]
- Tanifum EA, Starosolski ZA, Fowler S, Jankowsky JL, Annapragada AV. Cerebral vascular leak in a mouse model of amyloid neuropathology. J Cereb Blood Flow Metab. 2014 doi: 10.1038/jcbfm.2014.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tatebayashi Y, Miyasaka T, Chui DH, Akagi T, Mishima KI, Iwasaki K, Fujiwara M, Tanemura K, Murayama M, Ishiguro K, Planel E, Sato S, Hashikawa T, Takashima A. Tau filament formation and associative memory deficit in aged mice expressing mutant (R406W) human tau. Proceedings of the National Academy of Sciences. 2002;99:13896–13901. doi: 10.1073/pnas.202205599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toledo JB, Arnold SE, Raible K, Brettschneider J, Xie SX, Grossman M, Monsell S, Kukull W, Trojanowski JQ. Contribution of cerebrovascular disease in autopsy confirmed neurodegenerative disease cases in the National Alzheimer’s Coordinating Centre. Brain. 2013;136:2697–2706. doi: 10.1093/brain/awt188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsika E, Moysidou M, Guo J, Cushman M, Gannon P, Sandaltzopoulos R, Giasson BI, Krainc D, Ischiropoulos H, Mazzulli JR. Distinct region-specific alpha-synuclein oligomers in A53T transgenic mice: implications for neurodegeneration. J Neurosci. 2010;30:3409–18. doi: 10.1523/JNEUROSCI.4977-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verasdonck J, Bousset L, Gath J, Melki R, Böckmann A, Meier B. Further exploration of the conformational space of α-synuclein fibrils: solid-state NMR assignment of a high-pH polymorph. Biomolecular NMR Assignments. 2015:1–8. doi: 10.1007/s12104-015-9628-9. [DOI] [PubMed] [Google Scholar]
- Villarreal AE, Barron R, Rao KS, Britton GB. The Effects of Impaired Cerebral Circulation on Alzheimer’s Disease Pathology: Evidence from Animal Studies. Journal of Alzheimer’s Disease. 2014 doi: 10.3233/JAD-140144. [DOI] [PubMed] [Google Scholar]
- Violet M, Chauderlier A, Delattre L, Tardivel M, Chouala MS, Sultan A, Marciniak E, Humez S, Binder L, Kayed R, Lefebvre B, Bonnefoy E, Buée L, Galas M-C. Prefibrillar Tau oligomers alter the nucleic acid protective function of Tau in hippocampal neurons in vivo. Neurobiology of Disease. 2015;82:540–551. doi: 10.1016/j.nbd.2015.09.003. [DOI] [PubMed] [Google Scholar]
- Violet M, Delattre L, Tardivel M, Sultan A, Chauderlier A, Caillierez R, Talahari S, Nesslany F, Lefebvre B, Bonnefoy E, Buee L, Galas M-C. A major role for Tau in neuronal DNA and RNA protection in vivo under physiological and hyperthermic conditions. Frontiers in Cellular Neuroscience. 2014:8. doi: 10.3389/fncel.2014.00084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Von Bergen M, Barghorn S, Biernat J, Mandelkow E, Mandelkow E. Tau aggregation is driven by a transition from random coil to beta sheet structure. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2005;1739:158–166. doi: 10.1016/j.bbadis.2004.09.010. [DOI] [PubMed] [Google Scholar]
- Von Bergen M, Barghorn S, Li L, Marx A, Biernat J, Mandelkow EM, Mandelkow E. Mutations of Tau Protein in Frontotemporal Dementia Promote Aggregation of Paired Helical Filaments by Enhancing Local β-Structure. Journal of Biological Chemistry. 2001;276:48165–48174. doi: 10.1074/jbc.M105196200. [DOI] [PubMed] [Google Scholar]
- Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–9. doi: 10.1038/416535a. [DOI] [PubMed] [Google Scholar]
- Walsh DM, Lomakin A, Benedek GB, Condron MM, Teplow DB. Amyloid β-Protein Fibrillogenesis: DETECTION OF A PROTOFIBRILLAR INTERMEDIATE. Journal of Biological Chemistry. 1997;272:22364–22372. doi: 10.1074/jbc.272.35.22364. [DOI] [PubMed] [Google Scholar]
- Walsh DM, Selkoe DJ. Oligomers on the brain: the emerging role of soluble protein aggregates in neurodegeneration. Protein Pept Lett. 2004;11:213–28. doi: 10.2174/0929866043407174. [DOI] [PubMed] [Google Scholar]
- Walsh DM, Selkoe DJ. A beta oligomers - a decade of discovery. J Neurochem. 2007;101:1172–84. doi: 10.1111/j.1471-4159.2006.04426.x. [DOI] [PubMed] [Google Scholar]
- Wang Y, Martinez-Vicente M, Krüger U, Kaushik S, Wong E, Mandelkow EM, Cuervo AM, Mandelkow E. Tau fragmentation, aggregation and clearance: the dual role of lysosomal processing. Human Molecular Genetics. 2009;18:4153–4170. doi: 10.1093/hmg/ddp367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waxman EA, Giasson BI. Induction of intracellular tau aggregation is promoted by alpha-synuclein seeds and provides novel insights into the hyperphosphorylation of tau. J Neurosci. 2011;31:7604–18. doi: 10.1523/JNEUROSCI.0297-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wegmann S, Medalsy ID, Mandelkow E, Müller DJ. The fuzzy coat of pathological human Tau fibrils is a two-layered polyelectrolyte brush. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:E313–E321. doi: 10.1073/pnas.1212100110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams AD, Sega M, Chen M, Kheterpal I, Geva M, Berthelier V, Kaleta DT, Cook KD, Wetzel R. Structural properties of Aβ protofibrils stabilized by a small molecule. Proceedings of the National Academy of Sciences of the United States of America. 2005a;102:7115–7120. doi: 10.1073/pnas.0408582102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams S, Chalmers K, Wilcock GK, Love S. Relationship of neurofibrillary pathology to cerebral amyloid angiopathy in Alzheimer’s disease. Neuropathology and Applied Neurobiology. 2005b;31:414–421. doi: 10.1111/j.1365-2990.2005.00663.x. [DOI] [PubMed] [Google Scholar]
- Woerman AL, Stöhr J, Aoyagi A, Rampersaud R, Krejciova Z, Watts JC, Ohyama T, Patel S, Widjaja K, Oehler A, Sanders DW, Diamond MI, Seeley WW, Middleton LT, Gentleman SM, Mordes DA, Südhof TC, Giles K, Prusiner SB. Propagation of prions causing synucleinopathies in cultured cells. Proceedings of the National Academy of Sciences. 2015;112:E4949–E4958. doi: 10.1073/pnas.1513426112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu JW, Herman M, Liu L, Simoes S, Acker CM, Figueroa H, Steinberg JI, Margittai M, Kayed R, Zurzolo C, Di Paolo G, Duff KE. Small Misfolded Tau Species Are Internalized via Bulk Endocytosis and Anterogradely and Retrogradely Transported in Neurons. Journal of Biological Chemistry. 2013;288:1856–1870. doi: 10.1074/jbc.M112.394528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu L, Zheng J, Margittai M, Nussinov R, Ma B. Hyperphopsphorylation Promotes Tau Aggregation by Modulating Filament Structure and Stability. ACS Chemical Neuroscience. 2016 doi: 10.1021/acschemneuro.5b00294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yarchoan M, Xie S, Kling M, Toledo J, Wolk DA, Lee E, Van Deerlin V, Lee VMY, Trojanowski JQ, Arnold SE. Cerebrovascular atherosclerosis correlates with Alzheimer pathology in neurodegenerative dementias. Brain. 2012;135:3749–3756. doi: 10.1093/brain/aws271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshida M. Cellular tau pathology and immunohistochemical study of tau isoforms in sporadic tauopathies. Neuropathology. 2006;26:457–470. doi: 10.1111/j.1440-1789.2006.00743.x. [DOI] [PubMed] [Google Scholar]
- Zako T. Amyloid oligomers. FEBS J. 2010;277:1347. doi: 10.1111/j.1742-4658.2010.07567.x. [DOI] [PubMed] [Google Scholar]
- Zhao Y, Gu J, Dai C, Liu Q, Iqbal K, Liu F, Gong C. Chronic Cerebral Hypoperfusion Causes Decrease of O-GlcNAcylation, Hyperphosphorylation of Tau and Behavioral Deficits in Mice. Frontiers in Aging Neuroscience. 2014:6. doi: 10.3389/fnagi.2014.00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zilka N, Filipcik P, Koson P, Fialova L, Skrabana R, Zilkova M, Rolkova G, Kontsekova E, Novak M. Truncated tau from sporadic Alzheimer’s disease suffices to drive neurofibrillary degeneration in vivo. FEBS Letters. 2006;580:3582–3588. doi: 10.1016/j.febslet.2006.05.029. [DOI] [PubMed] [Google Scholar]



