Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 1.
Published in final edited form as: Biochim Biophys Acta Mol Cell Res. 2021 Feb 5;1868(5):118984. doi: 10.1016/j.bbamcr.2021.118984

Combating deleterious phase transitions in neurodegenerative disease

April L Darling 1, James Shorter 1,*
PMCID: PMC7965345  NIHMSID: NIHMS1670635  PMID: 33549703

Abstract

Protein aggregation is a hallmark of neurodegenerative diseases. However, the mechanism that induces pathogenic aggregation is not well understood. Recently, it has emerged that several of the pathological proteins found in an aggregated or mislocalized state in neurodegenerative diseases are also able to undergo liquid-liquid phase separation under physiological conditions. Although these phase transitions are important for various physiological functions, neurodegenerative disease-related mutations and conditions can alter the LLPS behavior of these proteins, which can elicit toxicity. Therefore, therapeutics that antagonize aberrant LLPS may be able to mitigate toxicity and aggregation that is ubiquitous in neurodegenerative disease. Here, we discuss the mechanisms by which aberrant protein phase transitions may contribute to neurodegenerative disease. We also outline potential therapeutic strategies to counter deleterious phases.

Keywords: liquid-liquid phase separation, RNA-binding proteins, intrinsically disordered proteins, FUS, TDP-43, tau, α-synuclein

Introduction

Cells are organized into compartments known as organelles. Conventionally, organelles were thought to be restricted to eukaryotes and were defined by a delimiting membrane that allows physiochemical separation from the rest of the cellular environment. However, it is now clear that organelles can also be formed without a membrane boundary. These organelles are termed membrane-less organelles (MLOs) and are found in eukaryotes and prokaryotes [1, 2]. MLOs form via spontaneous liquid-liquid phase separation (LLPS) of key biomolecules allowing them to achieve physiochemical separation from the surrounding environment. Some examples of eukaryotic MLOs include stress granules [3], nucleoli [4], P bodies [5], Cajal bodies [6], and paraspeckles [7].

In polymer physics, phase separation is a well-known phenomenon [8]. This phenomenon has also long been observed in structural biology during the process of protein crystallization [9], but is now appreciated anew in biology. In 2009, P granules were found to have liquid-like properties and form by phase separation [10]. In 2011, the nucleolus was also found to display liquid-like properties and forms via LLPS [11]. Several other instances of LLPS in biological systems have been defined, including both physiological and pathological examples. Here, we will describe pathological protein LLPS that is linked to neurodegenerative disease-related proteins. Further, we will outline potential therapeutic strategies to counter deleterious phases in neurodegenerative disease.

Common features of proteins that undergo LLPS

Proteins that undergo LLPS share common features. The most important feature that enables a protein to undergo LLPS is multivalency [1215]. Indeed, the concentration of protein needed to undergo LLPS is directly related to its valency, with a lower concentration needed for proteins with a higher valency [16]. Interestingly, several key neurotoxic proteins that are emblematic of neurodegenerative disease are also highly multivalent, and will readily undergo LLPS [17]. It has been suggested that LLPS may be a common feature of many proteins [18].

One way for a protein to realize its multivalency is to interact with other proteins in a dynamic network. Indeed, proteins that undergo LLPS also tend to have regions that lack a defined secondary structure, and this intrinsic disorder facilitates the flexibility to participate in a high number of multivalent interactions or, in other words, allows them to interact with multiple binding partners simultaneously. Thus, it is not surprising that many of the proteins that have been identified with LLPS behavior are intrinsically disordered [19]. It is important to note, however, that multivalency can also be achieved via folded domains [13]. Additionally, proteins with the capacity to oligomerize have a high valency and therefore are also common examples of proteins able to undergo LLPS. In fact, light-sensitive oligomerization domains from the protein cryptochrome-2 (Cry2) have been used to promote phase transitions optogenetically [2022]. Lastly, nucleic acid binding proteins have the potential to engage nucleic acid and protein partners, and this elevated level of valency makes them robust examples of LLPS-competent proteins [3, 2326]. Additionally, RNA can serve as a scaffold for LLPS to occur for several key RNA-binding proteins (RBPs), including those involved in neurodegenerative diseases such as FUS [27]. Generally, RNAs that serve as scaffolds are longer and have multiple binding sites that can dock several copies of the same RBP on one RNA molecule and therefore can nucleate the formation of higher order structures such as liquid droplets [15, 28]. Conversely, short RNAs lack the valency to make these higher-order contacts and therefore can antagonize LLPS [15, 21]. RNAs can also maintain proteins in a soluble state [2931]. Likewise, other multivalent scaffolds that engage RBPs, such as poly(ADP-ribose) can stimulate RBP LLPS as in the case of TDP-43 [32].

Pathological protein phase transitions in neurodegenerative diseases

Aberrant phase transitions are increasingly being associated with neurodegenerative disease pathology [3, 3344]. The role of pathological protein LLPS in disease is not well understood, but three potential mechanisms that have been proposed are illustrated in Figure 1. They include changes in the propensity of the protein to undergo LLPS (Figure 1A), changes to material properties of phase separated structures (Figure 1B), and nucleation of aggregation from sustained confinement in a restricted space that increases the probability for interaction (Figure 1C) [17].

Figure 1. Potential mechanisms by which aberrant liquid-liquid phase transitions might contribute to neurodegenerative diseases.

Figure 1.

LLPS is compared between a healthy neuron and a neuron that is deteriorating in neurodegenerative disease. (A) Disease-linked mutations or conditions can affect the ability of the protein to undergo LLPS, and in some cases the ability is enhanced while conversely in others it is diminished. (B) The material properties of a protein that has undergone LLPS may change due to disease-linked mutations or conditions. One such example is the liquid droplet losing ability to fuse. Another example of changes to material properties in a diseased neuron that is not shown is a loss of dynamics. (C) Sustained confinement in liquid droplets can lead to aggregation of the protein contents. This situation often arises for neurodegenerative disease-related proteins that pathologically aggregate. In a healthy neuron this scenario does not occur because LLPS is reversible and proteins in liquid droplets maintain dynamics. Figure made with Biorender.com.

Altered propensity to phase separate

Proteins that can undergo LLPS generally do so under specific conditions, as the process is extremely sensitive to changes in both the protein and environment [45]. This sensitivity may increase the possibility for dysregulation of LLPS-driven assemblies. Small perturbations can shift the phase boundary for a protein to undergo LLPS [46], and these shifts could have deleterious consequences for functional neuronal processes that rely on LLPS. There are several examples of these shifts occurring for proteins involved in neurodegenerative disease, and these examples will be discussed below.

Changes to material properties of phase separated droplets

When proteins undergo LLPS they form liquid droplets with high concentrations of the protein inside. While in a liquid state, these droplets possess certain material properties such as a high level of internal diffusion, the ability to fuse, and flow as a response to shear stress [10, 47]. However, with sustained time in a liquid droplet state, the proteins can convert to a more stable gel-like state, eventually converting into a solid with time [45]. One robust example is in the case of FUS [42]. FUS mutations underlie a subset of familial amyotrophic lateral sclerosis (ALS) cases [48, 49]. FUS rapidly undergoes LLPS, but over time converts to a less dynamic gel-like state, and eventually forms solid-like fibrillary aggregates [42, 5052]. Maturation of a liquid droplet into a solid over time is sometimes referred to as “molecular aging,” and could underlie pathological mechanisms that lead to neurodegenerative disease [41, 42]. Recent work also suggests that FUS can form soft glassy materials referred to as ‘Maxwell glasses’ with age-dependent material properties [53]. Therapeutics that focus on preserving the liquid-like properties of proteins that have undergone LLPS without allowing them to progress to other material states may be important for addressing this potential pathogenic mechanism.

Nucleation of aggregation

Neurodegenerative diseases share a common theme of protein misfolding leading to the appearance of protein aggregates in disease tissue [5456]. These aggregates are associated with cytotoxicity and lead to neuronal loss as well as cognitive and motor impairments [54, 55]. Elimination of pathological aggregates may serve as a therapeutic approach to treat neurodegenerative disease [5759]. However, the appearance of aggregates precedes disease symptoms and diagnosis [60]. Therefore, delineating the initial events leading to pathological aggregation could enable the identification of earlier therapeutic targets [60]. Aberrant protein LLPS may be an example of an early aggregation event that could be targeted. If proteins dwell for too long in a liquid-like state they risk conversion to pathological fibrils [3, 42]. Therefore, therapeutic agents could be developed to disaggregate or prevent the aggregation of proteins that have undergone LLPS [21, 57, 61].

Pathological proteins that undergo LLPS in ALS and frontotemporal dementia (FTD)

ALS is a progressive neurodegenerative disease that is characterized by the degeneration of upper and lower motor neurons [6264]. FTD is another progressive neurodegenerative disease that is characterized by degeneration of cortical neurons in the frontal and temporal lobes [65, 66]. ALS and FTD reside at opposite ends of a clinical spectrum of disorders, and patients can sometimes present with phenotypes of ALS and FTD [65, 66]. The pathological development of ALS/FTD is associated with aberrations in several key proteins including FUS, TDP-43, heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), ataxin 2, C9ORF72-derived dipeptide repeat proteins poly-GR and poly-PR, and ubiquilin-2 (UBQLN2), which, like in other neurodegenerative diseases, also appear as aggregates in diseased neurons [6769]. Which of these proteins undergoes aberrant phase transitions depends on the precise form of ALS/FTD [67, 70, 71]. Most notably, these proteins have all been identified as being components of stress granules, which are stress-induced MLOs containing various RNAs and proteins [67]. Stress granules are highly dynamic. They assemble when the cell is exposed to various stressors but dissipate once the stress subsides, and while they are formed, they readily exchange material with the cytosol [72]. Several links associate aberrant stress-granule assembly to disease pathogenesis, and these connections offer insight into potential therapeutics that may be promising for mitigating disease pathogenesis [22, 35, 36]. Next, we will focus on each of the proteins listed above, and examine how their LLPS may contribute to the development of ALS/FTD.

FUS

FUS is a predominantly nuclear RBP that is mislocalized to cytoplasmic inclusions in degenerating neurons that are hallmarks in ALS/FTD [67]. FUS associates with stress granules, and the first studies of FUS LLPS demonstrated that it can phase separate into spherical droplets able to fuse and recover after photobleaching, demonstrating their highly dynamic nature [42]. Structurally, FUS has an N-terminal prion-like domain (PrLD) [7375], which is necessary for its aggregation and LLPS behavior [15, 27, 51]. A FTD-linked FUS variant with a mutation in its PrLD, G156E, formed non-spherical structures that were unable to recover from photobleaching, indicating a change in material properties compared to wild-type (WT) FUS [42]. Additionally, ALS/FTD-linked mutations alter the mobility of phase-separated FUS in vitro and in vivo, and studies suggest that changing the material properties of FUS may contribute to disease [41]. One promising approach to modulate FUS phase behavior is through phosphorylation of the PrLD. In fact, when FUS is phosphorylated in its PrLD it is less prone to aggregation, shows a decreased toxicity in yeast models, and its phase-separation behavior is altered [76, 77]. Indeed, some phosphorylation events permit FUS LLPS but inhibit the pathological transition to gel and solid states [77].

TDP-43

TDP-43 is another predominantly nuclear RBP that is a major component of cytoplasmic inclusions found in degenerating neurons of ALS/FTD patients [67, 78]. TDP-43 proteinopathy and FUS proteinopathy are mutually exclusive in disease [70]. TDP-43 proteinopathy is also more common, being in found in ~97% of ALS cases compared to the ~1% of ALS cases that present with FUS proteinopathy [70]. In disease-linked inclusions, TDP-43 is often found as hyper-phosphorylated, ubiquitinated and cleaved C-terminal fragments [78]. TDP-43 has a C-terminal PrLD [7375], which is phosphorylated and aggregated in motor neurons of ALS patients and the majority of ALS-associated TDP-43 mutations are in this region [67, 79, 80]. The C-terminal PrLD renders TDP-43 intrinsically aggregation prone [81], and in isolation can phase separate into droplets with rapid dynamics as measured via fluorescence recovery after photobleaching [39]. Likewise, full-length TDP-43 can spontaneously undergo LLPS [15, 32, 82]. Some ALS-linked TDP-43 mutations accelerate aggregation at the expense of liquid states [21, 32, 39, 81]. Additionally, ALS-associated genes such as UBQLN2 and hnRNPA1 can alter TDP-43 aggregation in cell models [83].

A region within the TDP-43 PrLD spanning residues 320–343 appears critical for LLPS [39]. This region is evolutionarily conserved and exhibits a propensity to form an α-helix within the otherwise disordered PrLD [39]. Mutations that reduce α-helical propensity reduce TDP-43 LLPS, whereas mutations that enhance α-helical propensity increase LLPS, and can be ALS-linked as with G335D [84]. This region is required for TDP-43 LLPS in cells [85]. Intriguingly, perturbed LLPS affects TDP-43 splicing function at some exons but not others, indicating a nuanced role for LLPS in TDP-43 functionality in pre-mRNA splicing [84, 86].

Heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1)

hnRNPA1 is another predominantly nuclear RBP that can mislocalize to cytoplasmic inclusions in the degenerating tissues of multisystem proteinopathy (MSP) patients [35, 67, 87]. MSP patients sometimes present with motor neuron degeneration similar to ALS patients [63, 88]. hnRNPA1 has a C-terminal PrLD, and missense mutations in that region are linked to MSP and ALS [35]. For example, a D262V mutation introduces a potent steric zipper into the PrLD that leads to rapid hnRNPA1 fibrillization [35, 67]. hnRNAP1 also forms liquid droplets in vitro in a concentration-dependent and PrLD-dependent manner [3, 89]. Importantly, persistent droplet formation leads to aggregate accumulation within the droplet, which is aggravated by the D262V mutation [3, 89].

T-Cell restricted intracellular antigen-1 (TIA1)

TIA1 is a major protein component of stress granules. Condensation of its C-terminal PrLD may contribute to stress granule assembly [90]. Indeed, TIA1 undergoes spontaneous LLPS in the presence of RNA [89]. Disease-related mutations in the C-terminal PrLD (including P326L, A381T, and E384K) promote phase transitions, decrease dynamics and reversibility of droplets, and alter stress granule dynamics by inhibiting their rate of dissociation [91]. These properties likely combine to enhance the mislocalization and inclusion formation of TDP-43 [91].

Arginine-rich C9orf72-derived dipeptide repeats (poly-GR, poly-PR)

The most common genetic cause of ALS/FTD is a hexanucleotide repeat expansion in the gene C9orf72 [92, 93]. Although the expansion is found in a non-coding region of the gene, dipeptide repeats can be translated bidirectionally via repeat-associated non-AUG (RAN) translation, resulting in five distinct species: poly-GA, -GP, -GR, -PA, and -PR, as well as chimeric dipeptide fusions [69, 9496]. Although the toxic species and interactions of these dipeptide repeats continue to be unraveled, poly-GR and poly-PR have repeatedly been shown to be the most toxic in cellular and animal models [38, 97, 98]. Additionally, poly-GR and poly-PR rapidly phase separate in physiological conditions in the presence of RNA, and impair the assembly, dynamics, and function of MLOs [38, 69, 98100]. Poly-GR but not poly-GA also accelerate TDP-43 aggregation, which likely drives TDP-43 proteinopathy in c9ALS/FTD [68, 101].

Ataxin 2

Ataxin 2 contains a poly-Q tract in one of its three PrLDs, and when this expansion goes beyond a certain threshold (27–33 glutamines), it is implicated in ALS [102105]. By contrast, poly-Q tracts in ataxin 2 longer than 33 repeats are connected with spinocerebellar ataxia type 2 [106108]. An unbiased screen to identify modifiers of TDP-43 toxicity in yeast identified ataxin 2 as a deletion suppressor of TDP-43 toxicity [102]. TDP-43 and ataxin 2 associate in an RNA-dependent complex that mislocalizes to the cytoplasm in spinal cord neurons of ALS patients [102]. Ataxin 2 is also involved in the assembly of higher order MLOs known as mRNP assemblies that contribute to long-term memory and plasticity [33]. These assemblies are made up of RNP granules, which may act as vessels where pathogenic proteins aggregate [36, 74, 109]. Knockdown of ataxin 2 slows the progression of neurodegenerative pathologies connected to ALS with a targeted reduction in RNP granule formation efficiency [33, 110]. Indeed, antisense oligonucleotides that reduce ataxin 2 expression are now entering clinical trials for ALS (NCT04494256).

UBQLN2

UBQLN2 is implicated in the degradation of misfolded proteins through the ubiquitin-proteasome system and macroautophagy [111]. In some patients with ALS, it is found in cytosolic inclusions of degenerating motor neurons [112]. Several missense mutations in UBQLN2 are connected with ALS and all are found around an unusual Pxx repeat, which is present in UBQLN2 but not other UBQLNs [113]. Importantly, mutations in the Pxx region of UBQLN2 alter its LLPS behavior [34]. Additionally, Pxx mutants that enhanced the oligomerization of UBQLN2 decreased the concentration of the protein needed for LLPS and promoted solid-like assemblies [34]. These findings raise the question of whether ALS-associated mutations in the Pxx region potentially contribute to the aberrant stress granule dynamics, which may promote formation of the characteristic inclusions found in motor neurons of ALS patients.

Pathological proteins that undergo LLPS in Alzheimer’s disease

Alzheimer’s disease (AD) is a progressive fatal neurodegenerative disease with no cure or effective treatments [114]. The hallmarks of disease include extracellular amyloid-β plaques and intracellular tangles of aggregated tau [114]. Tau is a microtubule-associated protein and preventing its aggregation and spreading may be an important therapeutic approach to treat disease [115]. However, the initial events which seed the pathological aggregation of tau are not well understood. In addition to tau aggregation, there may also be a pathological contribution from RBP aggregates, which associate with tau aggregates in the brain of AD patients [116]. One such example is the RBP, U1 small nuclear ribonucleoprotein 70 kDa (U1–70K), but the co-aggregation mechanisms are incompletely understood [117].

Tau

Tau aggregation is a hallmark of AD and promotes neuronal loss and cognitive decline [118122]. Several other disorders are characterized by aberrant tau aggregation, which are referred to as tauopathies, one example of which is FTD [123, 124]. It has long been recognized that hyperphosphorylation of tau can cause it to dissociate from microtubules and specific tau phosphoforms can facilitate self-interaction and stimulate aggregation [125127]. However, the exact mechanism that causes tau to become pathological has remained enigmatic. Strategies to reduce tau aggregation and increase degradation of aberrant species can restore proteostasis and mitigate cognitive decline in AD mouse models [128, 129]. Tau can also undergo LLPS, which may help promote local nucleation of microtubule bundles [130]. However, tau LLPS can also initiate its aggregation and cause it to seed the aggregation of other proteins [44, 131133]. Phosphorylation can also increase the propensity of tau to undergo LLPS [44]. On the other hand, tau acetylation reduces tau LLPS [134].

U1–70K-RNA-binding protein (U1–70K)

U1–70K aggregates are also found in the brains of AD patients [117, 135]. Repetitive basic-acidic motifs located in two low-complexity domain (LCD) fragments of U1–70K induce phase separation, which leads to accelerated aggregation in vitro [136, 137]. Interestingly, whereas LLPS of many proteins depends on the presence of hydrophobic residues, U1–70K condenses in a charge-driven manner [136].

Pathological proteins that undergo LLPS found in Parkinson’s disease

Parkinson’s disease (PD) is the second most common neurodegenerative disease and is characterized by a progressive loss of dopaminergic neurons and accumulation of intracellular inclusions of aggregated α-synuclein [138]. The α-synuclein-rich inclusions, termed Lewy bodies, are associated with cytotoxicity and lead to neuronal loss and concomitant motor impairments [139147]. Prevention or elimination of pathological α-synuclein aggregates may serve as a therapeutic approach for PD treatment [148, 149].

α-Synuclein

α-Synuclein is a presynaptic protein with roles in neuronal survival and synaptic vesicle trafficking regulation [150]. Under physiological conditions in vitro, α-synuclein spontaneously forms amyloid fibrils, which are grossly similar to the amyloids found in Lewy bodies [151]. However, recent α-synuclein fibrils isolated from disease tissue can have distinct structures from those formed in vitro [152]. Preventing pathological α-synuclein aggregation rescues cells from death and restores cognitive and motor functions [153], but the exact mechanism leading to aggregation is not well understood. The LLPS behavior of α-synuclein may offer clues into how pathological aggregation is initiated. α-Synuclein undergoes spontaneous LLPS in the presence of molecular-crowding agents such as polyethylene glycol, and this process is highly dynamic and reversible at early stages [37]. However, sustained α-synuclein LLPS leads to a loss of dynamics and reversibility, and eventually aggregation [37]. Additionally, PD-related factors, including mutations (A53T, E46K, and S129E) and disease-relevant conditions, accelerate α-synuclein LLPS and subsequent fibril formation [37]. Thus, α-synuclein may populate liquid states prior to converting to fibrils in Lewy bodies. Indeed, in a C. elegans model of PD, α-synuclein forms liquid droplets that convert into amyloid-rich hydrogels, in a process that may mimic Lewy body assembly in PD [154]. Additionally, ultrastructural analyses of Lewy bodies from PD patient tissue indicates that they do not always contain cross-β fibrillar forms of α-synuclein [155157]. Thus, other higher-order forms of α-synuclein (such as liquid states) might contribute to the spontaneous assembly of Lewy bodies [37, 142, 154]. Lewy body formation that is seeded by preformed α-synuclein fibrils may bypass these liquid states or accelerate their conversion to solid states [144, 146].

Pathological proteins that undergo LLPS in Huntington’s disease

Huntington’s disease (HD) affects the central nervous system and is a fatal neurodegenerative disease. Within brain cells of patients, aggregates of a mutant form of polyQ-expanded huntingtin protein cause toxicity [158]. For the disease to occur, the length of the poly-Q tract must be beyond a threshold of 36 [159]. The length of the repeat expansion has a strong inverse correlation to the age of disease onset as well as disease severity [160]. Curiously, pathogenic polyQ expansions (range, 40–64 repeats) in huntingtin have also been found rare ALD/FTD cases that do not display any features of HD [161].

Huntingtin

Mutant polyQ-expanded fragments of huntingtin can undergo LLPS and form reversible liquid droplets [43]. The LLPS behavior is driven by the length of the polyQ tract and proline-rich regions in the protein, with a correlation between the length of the repeat and the ability to undergo LLPS [43]. Additionally, sustained population in the liquid state drives polyQ fibrillization [43].

Strategies to combat pathological protein phase transitions

Molecular chaperones and protein disaggregases

In mammalian cells, the abundance of proteins must be carefully regulated to ensure proteostasis [162]. Proteostasis is especially essential in neurons as they do not divide, and thus they are highly vulnerable to accumulation of toxic misfolded conformers that cannot be diluted out by cell division [163]. To maintain proteostasis, the cell depends on an extensive network of molecular chaperones and proteolytic systems [164]. There are over 150 molecular chaperones that come in many different forms. These chaperones assist with maintaining proteostasis through several mechanisms, including aiding in protein folding, protein degradation, translation of nascent peptides, and in helping misfolded proteins recover their native form [165]. In some specialized cases, molecular chaperones can possess protein-disaggregase activity where they recover natively folded proteins from aggregated species [54, 57, 61, 166168]. Several neurodegenerative diseases are associated with mutations in chaperone proteins suggesting that their normal function may be protective [169172]. Additionally, since neurodegenerative diseases are associated with an inability to maintain proteostasis, harnessing the power of molecular chaperones could be a promising therapeutic strategy [59, 61, 173176].

Several strategies are being developed to harness molecular chaperones and improve their activity so that they can be leveraged as effective therapeutics in neurodegenerative diseases. Many of these strategies could act as therapeutics to treat aberrant protein LLPS associated with neurodegenerative diseases. Below we will discuss the strategies for using protein disaggregases and other agents to treat aberrant LLPS that is characteristic in neurodegenerative diseases.

Potentiation of protein disaggregases

Molecular chaperones and protein disaggregases lose the battle against protein misfolding in the context of neurodegenerative disease [173]. One strategy to shift the balance toward protein folding has been to overexpress individual protein disaggregases or molecular chaperones, which can partially mitigate protein aggregation or neurodegenerative phenotypes in model systems [148, 149, 177181]. We have hypothesized that WT protein disaggregases may not be optimally adapted to reverse deleterious phase transitions in the environment of the aging brain [57, 174, 182]. Thus, we suggest that engineering potentiated protein disaggregases could be an effective strategy to increase their activity and specificity for their target clientele [57, 174, 182]. This strategy has been used primarily on the Hsp104 disaggregase to increase its activity to reverse pathological aggregation that is characteristic of neurodegenerative diseases [183185].

Hsp104 is a hexameric AAA+ ATPase and protein disaggregase found in all non-metazoan eukaryotes [59, 166, 186, 187]. It has successfully been engineered to have potentiated disaggregase activity [183185, 188]. Many genetically engineered versions of Hsp104 have now been shown to have enhanced activity and ability to revert TDP-43, FUS, and α-synuclein aggregation, suppress toxicity and eliminate protein aggregates in yeast [183185, 188].. Additionally, enhanced Hsp104 variants can dissolve cytoplasmic ALS-linked FUS and TDP-43 aggregates in mammalian cells [189, 190], and antagonize α-synuclein-mediated dopaminergic neurodegeneration in worm [188, 191].

While metazoan systems lack Hsp104, they do encode other AAA+ proteins that can function as protein disaggregases, including Skd3 [168] and VCP [192, 193]. Thus, Skd3 and VCP may also be candidates for engineering. Likewise, the human protein-disaggregase system, comprised of Hsp110, Hsp70, Hsp40, and small heat-shock proteins might also be targeted [174]. This system can disaggregate and reactivate proteins trapped in disordered aggregates and amyloid, but fails in the context of neurodegenerative disease [167, 194200]. Hsp70 also maintains liquid forms of TDP-43 in the nucleus and prevents their solidification [201]. The bacterial version of Hsp70 (DnaK) has been successfully engineered to outperform the WT version in refolding denatured proteins with an improved initial refolding rate [202]. Likewise, engineering efforts are also beginning to uncover mildly enhanced versions of human Hsp70 [174, 203205]. It will be interesting to define how potentiated disaggregases affect LLPS of protein clients, and discern if further genetic engineering could improve their ability to modulate LLPS.

Another human protein-disaggregase system that has recently come to light is comprised of the tripartite-motif (TRIM) proteins [149, 206]. TRIM proteins are defined by the TRIM/RBCC motif consisting of a RING domain, one or two B-boxes, and a coiled-coil region [207]. These proteins are found only in metazoans and exist as a large number of distinct variants, with ~20 in C. elegans and over 70 in mice and humans [207]. Recent studies have established that TRIM11 prevents the formation of protein aggregates and dissolves pre-existing protein deposits, including amyloid fibrils [149]. These molecular chaperone and disaggregase activities are ATP independent [149]. The chaperone and disaggregase activity of TRIM11 enhances the folding and solubility of normal proteins and cooperate with TRIM11 SUMO ligase activity to degrade aberrant proteins [149]. TRIM11 reverses α-synuclein fibrillization and mitigates α-synuclein toxicity in cell models of PD [149]. Intracranial delivery of TRIM11 mitigates α-synuclein-mediated pathology, neurodegeneration, and motor impairments in a PD mouse model [149]. Other TRIMs can also function as ATP-independent molecular chaperones and disaggregases, including TRIM19 and TRIM21 [149, 206]. Importantly, TRIM19 can be engineered to display enhanced activity against polyQ [208]. Thus, TRIMs are a potent and multifunctional protein quality-control system in metazoa, which might be applied and engineered to treat neurodegenerative diseases.

Conventional pharmacological treatments have proven difficult to develop for the treatment of neurodegenerative diseases. This difficulty reflects, at least in part, the challenges for drugs to cross the blood-brain barrier and the risk of direct administration to the central nervous system. One approach that has been successfully conceptualized for the treatment of such disorders is through gene transfer mediated by an adeno-associated virus (AAV) [209]. After a single administration of AAV, non-dividing cells like neurons can be successfully transduced allowing for the permanent expression of a therapeutic gene of interest [210]. The safety of this delivery method has been consistently demonstrated in clinical trials for neurodegenerative diseases [211213], but most transgenes have shown limited efficacy. This limited efficacy highlights the need for a transgene that can address the root underlying cause of the disease. Protein disaggregases such as TRIMs or engineered variants may be promising targets for delivery by AAV [149, 206208].

Drugs that upregulate chaperone expression

Another possible method to induce the expression of chaperones or disaggregases is through stimulation by brain-penetrant small-molecule drugs. Here, rather than overexpressing an individual disaggregase or enhanced variants, the strategy is to elicit transcription of a battery of chaperones or specific chaperones as might occur in a stress response [214, 215]. However, this strategy assumes that transcriptional stress responses can still occur productively under the conditions of neurodegenerative disease or aging, which may not be the case [214, 216, 217]. Likewise, chronic overactivation of transcriptional stress responses can also be damaging, and there may also be merit in inhibiting their overactivation [218223]. Nonetheless, there have been several drugs that have been developed for boosting chaperone expression and some are undergoing clinical trials for treatment of ALS [224], including arimoclomal and colchicine.

Arimoclomol is an analog of bimoclomol, a compound with stress protein-inducing activity and cytoprotective effects that is non-toxic [225, 226]. Specifically, arimoclomol is a co-inducer of Hsp70 and Hsp90, and it is able to delay disease progression in ALS and inclusion body myopathy mice [227, 228]. Moreover, a randomized, double-blind, placebo-controlled trial of arimoclomol treatment in rapidly progressive SOD1 ALS patients concluded that the drug was safe and well tolerated, but not powerful enough for a therapeutic effect [229]. However, it was noted that a larger sample size may show a therapeutic effect because a positive, but not significant, trend was observed [229]. Furthermore, the identification of superior biomarkers that can be used to diagnose ALS earlier may also improve the prospects for arimoclomol. Earlier administration of arimoclomol may enable greater neuroprotection.

Colchicine is a microtubule-destabilizing drug that leads to an upregulation of a chaperone protein. Colchicine greatly enhances expression of HspB8, a small heat shock protein, as well as the expression of several other proteins involved in autophagy [230]. In neurons, the expression of HspB8 is normally induced when proteasomal impairments occur [231, 232]. HspB8 is also part of the Hsp70-Bag3-HspB8 complex. This complex recognizes and responds to misfolded proteins in stress granules and removes them for refolding or degradation, which facilitates their clearance [233]. Interestingly, HspB8 mRNA levels are significantly higher in the spinal cord of ALS patients than in age-matched controls, leading to the hypothesis that its expression is involved in the cellular response to molecular insults associated with ALS [234]. Indeed, when HspB8 is induced by colchicine, it facilitates the removal of insoluble accumulations of TDP-43 [235]. Moreover, in a transgenic ALS SOD1-G93A mouse model, there was a robust increase of HspB8 protein levels in the anterior horn cells of the spinal cord that survived to end-stage of disease [232]. This increase was associated with more diffuse, non-aggregated mutant SOD1 [232]. The effects of upregulating HspB8 expression with colchicine may be promising, and there is an ongoing phase II multicenter clinical trial to assess colchicine as a potential therapeutic for ALS (NCT03693781).

Nuclear-import receptors (NIRs)

When molecules pass from the cytoplasm to the nucleus, they are transported through the nuclear pore complex [236]. Small molecules are able to pass through via passive transport, but large proteins (>20kDa) must usually engage nuclear-import receptors (NIRs) via a nuclear localization signal (NLS) to be effectively transported across the nuclear pore complex [236]. NIRs are typically members of the Karyopherin-β (Kap-β) family, which drive most cytoplasm-to-nucleus transport events [236].

NIRs have been shown to drive nuclear localization of RBPs that are connected with neurodegenerative disease pathology [237] and, importantly, can chaperone and disaggregate disease-linked RBPs with an NLS [25, 52, 101, 238243]. In fact, by binding to the NLS of its cargo, Karyopherin-β2 (Kapβ2) can prevent and reverse fibrillization of FUS, hnRNPA1, hnRNPA2, TAF15, EWSR1, and several disease-linked variants [52]. Kapβ2 can also prevent poly-GR from stimulating TDP-43 aggregation, and NIRs can also directly antagonize poly-GR condensation [101]. Moreover, importin α and Kapβ1 prevent and reverse TDP-43 condensation and fibrillization [52, 101]. Nuclear magnetic resonance spectroscopy suggested that after Kapβ2 binds the NLS of FUS it then engages with sequence elements and structural domains distributed throughout FUS that contribute to FUS phase separation [243]. Thus, the chaperone activity of Kapβ2 is achieved without ATP hydrolysis, and occurs by competing with multivalent cargo-cargo interactions that facilitate oligomerization [243]. Hence, Kapβ2 and other NIRs prevent aberrant accumulation of RBPs containing NLSs into cytoplasmic inclusions, and re-establish their nuclear localization and function, rescuing the degeneration caused by mislocalized RBPs in disease [52, 61]. Identifying small-molecule drugs to increase expression or stimulate NIR activity (e.g. by stabilizing NIR interactions with specific cargo such as TDP-43 or FUS) is another potential approach to treating diseases associated with aberrant phase transitions [52, 61].

RNA and oligonucleotides

As a reciprocal strategy to increasing chaperone activity, anti-sense oligonucleotides (ASOs) to modulate expression of specific proteins represent attractive therapeutic strategies in treating neurological diseases [56, 244246]. Indeed, ASOs are now approved therapeutics for spinal muscular atrophy [56, 244]. Several possibilities for ASO interventions in neurodegenerative disease are apparent. For example, TIA1 is a key protein involved in stress granule assembly [91]. It also has been shown to interact with tau, and interestingly, knockdown of TIA1 via ASOs prevented tau pathology and toxicity in primary neurons and AD rodent models [247, 248]. Another example of successful treatment by ASOs is through the knock-down of ataxin 2, another key protein involved in the regulation of stress granules [110]. Targeting ataxin 2 via ASOs led to reduced TDP-43 pathology in a transgenic mouse model of TDP-43 proteinopathy and increased survival [110]. Additionally, the deletion of a key region of ataxin 2 involved in LLPS prevented neurodegeneration induced by both FUS and c9orf72 dipeptide repeats [33]. Ataxin 2 ASOs are now in phase 1/2 clinical trials (NCT04494256). Likewise, several other neurodegenerative disease proteins are being targeted by therapeutic ASOs, including SOD1 [245, 249], huntingtin [250], prion protein [251, 252], α-synuclein [253, 254], and c9orf72 [245, 255257]. In these cases, it is thought that knockdown of these genes will not have damaging side effects. The situation is more complex for FUS and TDP-43, which serve more essential functions, because reduced expression could have negative effects on neurons [70]. However, in some genetic forms of disease it may still be possible to specifically knockdown a mutant form of TDP-43 (e.g. M337V) [258] or FUS (e.g. P525L) [259] which is causing disease. Here, selectively knocking down the mutant but not WT version of the RBP may be therapeutic.

More recently it has been shown that RNAs themselves can act as chaperones to prevent aberrant phase transitions [99, 260, 261]. Indeed, many of the pathological proteins involved in neurodegenerative disorders (particularly in ALS) are RBPs, and their propensity for self-association varies as a function of the RNA in their environment [260]. For instance, abnormal interactions between residues in the PrLD of TDP-43 facilitates its pathological accumulation [81, 262]. However, in the presence of RNA, this self-interaction is antagonized, while mutations in the RNA-recognition motifs of TDP-43 that reduce RNA binding also ablate the protective function of RNA [21]. The chaperone activity of RNA occurs both in vitro and in cells, as aberrant and toxic LLPS of TDP-43 in the cytoplasm of neurons could be rescued with an intervention of oligonucleotides designed to bind to TDP-43 RRMs [21]. This “bait RNA” was able to prevent aberrant TDP-43 inclusions and delay cell death in multiple cell models [21]. These results beg the question as to whether this bait RNA strategy can be used for other RBPs [263, 264].

Anti-Aggregation molecules

Small molecules that specifically bind to and prevent the aggregation of disease-linked proteins may be promising targets for the intervention of aberrant LLPS. One such example is AIM4, an acridine derivative, which interacts with the C-terminal PrLD of TDP-43 and prevents its aggregation [265]. Akin to the effects of RNA on TDP-43 LLPS, AIM4 can inhibit LLPS of a C-terminal TDP-43 fragment bearing an A315T familial ALS mutation [265]. Therefore, anti-aggregation molecules that prevent aggregation of their target proteins by binding to regions also important for their LLPS may be promising therapeutic leads. More studies are needed to explore the effects of other small molecules with this mechanism of action. Often an aggregation-prone protein will escape inhibition by a single small-molecule inhibitor by forming alternative aggregated structures or ‘strains’ [266268]. However, this process can be mitigated by combinations of two or more small molecules that synergize to antagonize aggregation [266268]. Thus, it will also be of great interest to find small-molecule combinations that synergize to antagonize aberrant LLPS to ensure maximal inhibition and minimize formation of alternative structural polymorphs.

Modulating ATP levels

As humans age, the risk of being diagnosed with a neurodegenerative disease substantially increases. Aging is also accompanied by a reduction in ATP levels [269]. This ATP depletion has an effect on cellular LLPS, as lowering cellular ATP levels decreases the dynamics of stress granules and makes them less reversible [270]. This loss of reversibility is most likely due to the fact that stress-granule disassembly is an ATPase-driven process [193]. Additionally, enhanced levels of ATP in the cell can act as a chemical hydrotrope that directly prevents phase separation and subsequent aggregation [271]. Therefore, strategies to increase the levels of ATP in cells may also be effective at combatting aberrant LLPS that is observed in neurodegenerative diseases.

Alteration of protein post-translational modifications

Post-translation modifications (PTMs) of proteins can have large effects on their activity and structure, including altering protein charge, size, and hydrophobicity. PTMs occur via the addition of functional groups by specialized enzymes or through more subtle changes such as oxidation [272]. Many protein PTMs serve as on/off switches, which regulate cellular processes [273] and LLPS [274]. There are several examples of PTMs on neurodegenerative disease-associated proteins that can alter both their aggregation characteristics as well as their LLPS behavior. Although several PTMs are associated with changes in protein LLPS, a potent example is phosphorylation.

Protein phosphorylation is the process of covalently adding a phosphoryl group to a hydroxyl group of a polar uncharged or negatively charged amino acid residue. The most common residues to be phosphorylated are serine, threonine, and tyrosine [275]. The addition of a phosphate group adds an additional charge to the protein, which can promote different interactions that, depending on their location, can either promote or inhibit LLPS. It is possible that phosphorylation will promote charge-charge interactions that drive LLPS [276], but alternatively it could cause charge repulsion or steric hindrance which instead inhibits LLPS [277].

The phosphorylation of serine/threonine residues in TIA1 [278] and tau [44] promote their LLPS behavior. Interestingly, phosphorylation of these proteins at the same residues that promote LLPS behavior also promotes their aggregation [44, 278]. Additionally, hyperphosphorylation of tau is a key biomarker in the AD patient brain [279], and specific tau phosphoforms exhibit accelerated fibrillization [126, 127].

By contrast, phosphorylation of the PrLD of FUS reduces LLPS [280]. In fact, FUS aggregation and LLPS are diminished when the FUS PrLD is phosphorylated or bears phosphomimetic substitutions [76, 77, 281]. Phosphorylation of the FUS PrLD introduces electrostatic charges that reduce its prion-like nature and multivalent interactions that drive LLPS [76]. Phosphorylation of the TDP-43 PrLD significant decreases in the number of cells containing TDP-43 puncta [282]. However, the phase separation of TDP-43 is also regulated PTMs in its N-terminal structured domain. Phosphorylation of serine 48 in the N-terminal domain, which is highly conserved across species, is enough to suppress TDP-43 LLPS [82].

Collectively, these findings suggest that genetic or pharmacological intervention to upregulate or downregulate specific PTMs may be an attractive therapeutic strategy against aberrant LLPS. However, before this strategy can be realized the target protein in question must be well characterized regarding how PTMs affect its LLPS behavior, propensity to aggregate, and toxicity.

Conclusions

It is now established that several of the pathological proteins found in an aggregated or mislocalized state in neurodegenerative diseases are also able to undergo LLPS under physiological conditions. Although these phase transitions are important for various physiological functions, neurodegenerative disease-related mutations and conditions can alter the LLPS behavior of these proteins, which can elicit toxicity. Thus, therapeutics that antagonize aberrant LLPS may be able to mitigate toxicity and aggregation that is ubiquitous in neurodegenerative disease. Here, we have discussed the mechanisms by which aberrant protein phase transitions may contribute to neurodegenerative disease. We have also outlined several potential therapeutic strategies to counter deleterious phases. We anticipate that these strategies alone or perhaps in synergistic combination will provide a powerful arsenal to ultimately eliminate neurodegenerative disease.

Highlights.

  • Discussion of deleterious phase transitions in neurodegenerative disease

  • Discussion of therapeutic strategies to counter deleterious phase transitions

  • Discussion of protein-disaggregase technologies as therapeutic agents

Acknowledgments

We thank Charlotte Fare, Katie Copley, Bede Portz, and Hana Odeh for critiques. A.L.D. is supported by a post-doctoral fellowship from the American Parkinson Disease Association. J.S. is supported by grants from NIH (R01GM099836, R21AG061784, and R21AG065854), Target ALS, the ALS Association, the Packard Center for ALS Research, the G. Harold & Leila Y. Mathers Foundation, Sanofi, and the Office of the Assistant Secretary of Defense for Health Affairs, through the Amyotrophic Lateral Sclerosis Research Program under award no. W81XWH-17-1-0237.

List of abbreviations

AAV

Adeno associated virus

AD

Alzheimer’s disease

ALS

Amyotrophic Lateral Sclerosis

ASO

Anti-sense oligonucleotide

Cry2

Cytochrome 2

FTD

Frontotemporal dementia

FUS

Fused in sarcoma

hnRNPA1

Heterogeneous nuclear ribonucleoprotein A1

HD

Huntington’s disease

Kapβ

Karyopherin-β

LLPS

Liquid-liquid phase separation

LCD

Low-complexity domain

MLO

Membrane-less organelles

Tau

Microtubule associated protein tau

NIR

Nuclear-import receptor

NLS

Nuclear-localization signal

PolyQ

Poly-glutamine

PD

Parkinson’s disease

PTM

Post translational modification

PrLD

Prion-like domain

RAN

Repeat associated non-AUG

RBP

RNA-binding protein

SCA3

Spinocerebellar ataxia type 3

TDP-43

TAR-DNA binding protein 43kDa

TIA1

T-Cell restricted intracellular antigen-1

U1–70K

U1 small nuclear ribonucleoprotein 70 kDa

UBQLN2

Ubiquilin 2

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of interests

A.L.D. has nothing to declare. J.S. is a consultant for Dewpoint Therapeutics and Maze Therapeutics.

References

  • [1].Azaldegui CA, Vecchiarelli AG, Biteen JS, The Emergence of Phase Separation as an Organizing Principle in Bacteria, Biophys J, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Lyon AS, Peeples WB, Rosen MK, A framework for understanding the functions of biomolecular condensates across scales, Nat Rev Mol Cell Biol, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Molliex A, Temirov J, Lee J, Coughlin M, Kanagaraj AP, Kim HJ, Mittag T, Taylor JP, Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization, Cell, 163 (2015) 123–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Feric M, Vaidya N, Harmon TS, Mitrea DM, Zhu L, Richardson TM, Kriwacki RW, Pappu RV, Brangwynne CP, Coexisting Liquid Phases Underlie Nucleolar Subcompartments, Cell, 165 (2016) 1686–1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Kroschwald S, Maharana S, Mateju D, Malinovska L, Nuske E, Poser I, Richter D, Alberti S, Promiscuous interactions and protein disaggregases determine the material state of stress-inducible RNP granules, Elife, 4 (2015) e06807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Neugebauer KM, Special focus on the Cajal Body, RNA Biol, 14 (2017) 669–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Nishimoto Y, Nakagawa S, Hirose T, Okano HJ, Takao M, Shibata S, Suyama S, Kuwako K, Imai T, Murayama S, Suzuki N, Okano H, The long non-coding RNA nuclear-enriched abundant transcript 1_2 induces paraspeckle formation in the motor neuron during the early phase of amyotrophic lateral sclerosis, Mol Brain, 6 (2013) 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Flory PJ, Thermodynamics of High Polymer Solutions, The Journal of Chemical Physics, 10 (1942) 51–61. [Google Scholar]
  • [9].Lomakin A, Asherie N, Benedek GB, Liquid-solid transition in nuclei of protein crystals, Proc Natl Acad Sci U S A, 100 (2003) 10254–10257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Brangwynne CP, Eckmann CR, Courson DS, Rybarska A, Hoege C, Gharakhani J, Jülicher F, Hyman AA, Germline P granules are liquid droplets that localize by controlled dissolution/condensation, Science, 324 (2009) 1729–1732. [DOI] [PubMed] [Google Scholar]
  • [11].Brangwynne CP, Mitchison TJ, Hyman AA, Active liquid-like behavior of nucleoli determines their size and shape in Xenopus laevis oocytes, Proc Natl Acad Sci U S A, 108 (2011) 4334–4339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Hyman AA, Weber CA, Jülicher F, Liquid-liquid phase separation in biology, Annu Rev Cell Dev Biol, 30 (2014) 39–58. [DOI] [PubMed] [Google Scholar]
  • [13].Martin EW, Holehouse AS, Intrinsically disordered protein regions and phase separation: sequence determinants of assembly or lack thereof, Emerg Top Life Sci, (2020). [DOI] [PubMed] [Google Scholar]
  • [14].Gomes E, Shorter J, The molecular language of membraneless organelles, J Biol Chem, 294 (2019) 7115–7127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Portz B, Lee BL, Shorter J, FUS and TDP-43 Phases in Health and Disease, Trends Biochem Sci, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Li P, Banjade S, Cheng H-C, Kim S, Chen B, Guo L, Llaguno M, Hollingsworth JV, King DS, Banani SF, Russo PS, Jiang Q-X, Nixon BT, Rosen MK, Phase transitions in the assembly of multivalent signalling proteins, Nature, 483 (2012) 336–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Elbaum-Garfinkle S, Matter over mind: Liquid phase separation and neurodegeneration, The Journal of biological chemistry, 294 (2019) 7160–7168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Hardenberg M, Horvath A, Ambrus V, Fuxreiter M, Vendruscolo M, Widespread occurrence of the droplet state of proteins in the human proteome, Proc Natl Acad Sci U S A, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Uversky VN, Supramolecular Fuzziness of Intracellular Liquid Droplets: Liquid-Liquid Phase Transitions, Membrane-Less Organelles, and Intrinsic Disorder, Molecules, 24 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Shin Y, Berry J, Pannucci N, Haataja MP, Toettcher JE, Brangwynne CP, Spatiotemporal Control of Intracellular Phase Transitions Using Light-Activated optoDroplets, Cell, 168 (2017) 159–171.e114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Mann JR, Gleixner AM, Mauna JC, Gomes E, DeChellis-Marks MR, Needham PG, Copley KE, Hurtle B, Portz B, Pyles NJ, Guo L, Calder CB, Wills ZP, Pandey UB, Kofler JK, Brodsky JL, Thathiah A, Shorter J, Donnelly CJ, RNA Binding Antagonizes Neurotoxic Phase Transitions of TDP-43, Neuron, 102 (2019) 321–338.e328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Zhang P, Fan B, Yang P, Temirov J, Messing J, Kim HJ, Taylor JP, Chronic optogenetic induction of stress granules is cytotoxic and reveals the evolution of ALS-FTD pathology, Elife, 8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Bogaert E, Boeynaems S, Kato M, Guo L, Caulfield TR, Steyaert J, Scheveneels W, Wilmans N, Haeck W, Hersmus N, Molecular dissection of FUS points at synergistic effect of low-complexity domains in toxicity, Cell reports, 24 (2018) 529–537. e524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Elbaum-Garfinkle S, Kim Y, Szczepaniak K, Chen CC-H, Eckmann CR, Myong S, Brangwynne CP, The disordered P granule protein LAF-1 drives phase separation into droplets with tunable viscosity and dynamics, Proceedings of the National Academy of Sciences, 112 (2015) 7189–7194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Qamar S, Wang G, Randle SJ, Ruggeri FS, Varela JA, Lin JQ, Phillips EC, Miyashita A, Williams D, Ströhl F, FUS phase separation is modulated by a molecular chaperone and methylation of arginine cation-π interactions, Cell, 173 (2018) 720–734. e715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Wang J, Choi J-M, Holehouse AS, Lee HO, Zhang X, Jahnel M, Maharana S, Lemaitre R, Pozniakovsky A, Drechsel D, A molecular grammar governing the driving forces for phase separation of prion-like RNA binding proteins, Cell, 174 (2018) 688–699. e616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Burke KA, Janke AM, Rhine CL, Fawzi NL, Residue-by-residue view of in vitro FUS granules that bind the C-terminal domain of RNA polymerase II, Molecular cell, 60 (2015) 231–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Schwartz JC, Wang X, Podell ER, Cech TR, RNA seeds higher-order assembly of FUS protein, Cell reports, 5 (2013) 918–925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Aarum J, Cabrera CP, Jones TA, Rajendran S, Adiutori R, Giovannoni G, Barnes MR, Malaspina A, Sheer D, Enzymatic degradation of RNA causes widespread protein aggregation in cell and tissue lysates, EMBO Rep, 21 (2020) e49585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Horowitz S, Bardwell JC, RNAs as chaperones, RNA Biol, 13 (2016) 1228–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Begeman A, Son A, Litberg TJ, Wroblewski TH, Gehring T, Huizar Cabral V, Bourne J, Xuan Z, Horowitz S, G-Quadruplexes act as sequence-dependent protein chaperones, EMBO Rep, 21 (2020) e49735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].McGurk L, Gomes E, Guo L, Mojsilovic-Petrovic J, Tran V, Kalb RG, Shorter J, Bonini NM, Poly(ADP-Ribose) Prevents Pathological Phase Separation of TDP-43 by Promoting Liquid Demixing and Stress Granule Localization, Mol Cell, 71 (2018) 703–717 e709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Bakthavachalu B, Huelsmeier J, Sudhakaran IP, Hillebrand J, Singh A, Petrauskas A, Thiagarajan D, Sankaranarayanan M, Mizoue L, Anderson EN, Pandey UB, Ross E, VijayRaghavan K, Parker R, Ramaswami M, RNP-Granule Assembly via Ataxin-2 Disordered Domains Is Required for Long-Term Memory and Neurodegeneration, Neuron, 98 (2018) 754–766.e754. [DOI] [PubMed] [Google Scholar]
  • [34].Dao TP, Martyniak B, Canning AJ, Lei Y, Colicino EG, Cosgrove MS, Hehnly H, Castañeda CA, ALS-Linked Mutations Affect UBQLN2 Oligomerization and Phase Separation in a Position- and Amino Acid-Dependent Manner, Structure, 27 (2019) 937–951.e935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Kim HJ, Kim NC, Wang YD, Scarborough EA, Moore J, Diaz Z, MacLea KS, Freibaum B, Li S, Molliex A, Kanagaraj AP, Carter R, Boylan KB, Wojtas AM, Rademakers R, Pinkus JL, Greenberg SA, Trojanowski JQ, Traynor BJ, Smith BN, Topp S, Gkazi AS, Miller J, Shaw CE, Kottlors M, Kirschner J, Pestronk A, Li YR, Ford AF, Gitler AD, Benatar M, King OD, Kimonis VE, Ross ED, Weihl CC, Shorter J, Taylor JP, Mutations in prion-like domains in hnRNPA2B1 and hnRNPA1 cause multisystem proteinopathy and ALS, Nature, 495 (2013) 467–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Li YR, King OD, Shorter J, Gitler AD, Stress granules as crucibles of ALS pathogenesis, J Cell Biol, 201 (2013) 361–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Ray S, Singh N, Kumar R, Patel K, Pandey S, Datta D, Mahato J, Panigrahi R, Navalkar A, Mehra S, Gadhe L, Chatterjee D, Sawner AS, Maiti S, Bhatia S, Gerez JA, Chowdhury A, Kumar A, Padinhateeri R, Riek R, Krishnamoorthy G, Maji SK, α-Synuclein aggregation nucleates through liquid-liquid phase separation, Nat Chem, (2020). [DOI] [PubMed] [Google Scholar]
  • [38].White MR, Mitrea DM, Zhang P, Stanley CB, Cassidy DE, Nourse A, Phillips AH, Tolbert M, Taylor JP, Kriwacki RW, C9orf72 Poly(PR) Dipeptide Repeats Disturb Biomolecular Phase Separation and Disrupt Nucleolar Function, Mol Cell, 74 (2019) 713–728.e716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Conicella AE, Zerze GH, Mittal J, Fawzi NL, ALS Mutations Disrupt Phase Separation Mediated by alpha-Helical Structure in the TDP-43 Low-Complexity C-Terminal Domain, Structure, 24 (2016) 1537–1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Dao TP, Kolaitis RM, Kim HJ, O’Donovan K, Martyniak B, Colicino E, Hehnly H, Taylor JP, Castaneda CA, Ubiquitin Modulates Liquid-Liquid Phase Separation of UBQLN2 via Disruption of Multivalent Interactions, Mol Cell, 69 (2018) 965–978.e966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Murakami T, Qamar S, Lin JQ, Schierle GS, Rees E, Miyashita A, Costa AR, Dodd RB, Chan FT, Michel CH, Kronenberg-Versteeg D, Li Y, Yang SP, Wakutani Y, Meadows W, Ferry RR, Dong L, Tartaglia GG, Favrin G, Lin WL, Dickson DW, Zhen M, Ron D, Schmitt-Ulms G, Fraser PE, Shneider NA, Holt C, Vendruscolo M, Kaminski CF, St George-Hyslop P, ALS/FTD Mutation-Induced Phase Transition of FUS Liquid Droplets and Reversible Hydrogels into Irreversible Hydrogels Impairs RNP Granule Function, Neuron, 88 (2015) 678–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Patel A, Lee HO, Jawerth L, Maharana S, Jahnel M, Hein MY, Stoynov S, Mahamid J, Saha S, Franzmann TM, Pozniakovski A, Poser I, Maghelli N, Royer LA, Weigert M, Myers EW, Grill S, Drechsel D, Hyman AA, Alberti S, A Liquid-to-Solid Phase Transition of the ALS Protein FUS Accelerated by Disease Mutation, Cell, 162 (2015) 1066–1077. [DOI] [PubMed] [Google Scholar]
  • [43].Peskett TR, Rau F, O’Driscoll J, Patani R, Lowe AR, Saibil HR, A Liquid to Solid Phase Transition Underlying Pathological Huntingtin Exon1 Aggregation, Mol Cell, 70 (2018) 588–601.e586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Wegmann S, Eftekharzadeh B, Tepper K, Zoltowska KM, Bennett RE, Dujardin S, Laskowski PR, MacKenzie D, Kamath T, Commins C, Vanderburg C, Roe AD, Fan Z, Molliex AM, Hernandez-Vega A, Muller D, Hyman AA, Mandelkow E, Taylor JP, Hyman BT, Tau protein liquid-liquid phase separation can initiate tau aggregation, Embo j, 37 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Boeynaems S, Alberti S, Fawzi NL, Mittag T, Polymenidou M, Rousseau F, Schymkowitz J, Shorter J, Wolozin B, Van Den Bosch L, Tompa P, Fuxreiter M, Protein Phase Separation: A New Phase in Cell Biology, Trends Cell Biol, 28 (2018) 420–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Cinar H, Fetahaj Z, Cinar S, Vernon RM, Chan HS, Winter RHA, Temperature, Hydrostatic Pressure, and Osmolyte Effects on Liquid-Liquid Phase Separation in Protein Condensates: Physical Chemistry and Biological Implications, Chemistry, 25 (2019) 13049–13069. [DOI] [PubMed] [Google Scholar]
  • [47].Kasza KE, Rowat AC, Liu J, Angelini TE, Brangwynne CP, Koenderink GH, Weitz DA, The cell as a material, Curr Opin Cell Biol, 19 (2007) 101–107. [DOI] [PubMed] [Google Scholar]
  • [48].Kwiatkowski TJ Jr., Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, Davis A, Gilchrist J, Kasarskis EJ, Munsat T, Valdmanis P, Rouleau GA, Hosler BA, Cortelli P, de Jong PJ, Yoshinaga Y, Haines JL, Pericak-Vance MA, Yan J, Ticozzi N, Siddique T, McKenna-Yasek D, Sapp PC, Horvitz HR, Landers JE, Brown RH Jr., Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis, Science, 323 (2009) 1205–1208. [DOI] [PubMed] [Google Scholar]
  • [49].Vance C, Rogelj B, Hortobágyi T, De Vos KJ, Nishimura AL, Sreedharan J, Hu X, Smith B, Ruddy D, Wright P, Ganesalingam J, Williams KL, Tripathi V, Al-Saraj S, Al-Chalabi A, Leigh PN, Blair IP, Nicholson G, de Belleroche J, Gallo JM, Miller CC, Shaw CE, Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6, Science, 323 (2009) 1208–1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Kato M, Han TW, Xie S, Shi K, Du X, Wu LC, Mirzaei H, Goldsmith EJ, Longgood J, Pei J, Grishin NV, Frantz DE, Schneider JW, Chen S, Li L, Sawaya MR, Eisenberg D, Tycko R, McKnight SL, Cell-free formation of RNA granules: low complexity sequence domains form dynamic fibers within hydrogels, Cell, 149 (2012) 753–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Sun Z, Diaz Z, Fang X, Hart MP, Chesi A, Shorter J, Gitler AD, Molecular determinants and genetic modifiers of aggregation and toxicity for the ALS disease protein FUS/TLS, PLoS Biol, 9 (2011) e1000614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Guo L, Kim HJ, Wang H, Monaghan J, Freyermuth F, Sung JC, O’Donovan K, Fare CM, Diaz Z, Singh N, Zhang ZC, Coughlin M, Sweeny EA, DeSantis ME, Jackrel ME, Rodell CB, Burdick JA, King OD, Gitler AD, Lagier-Tourenne C, Pandey UB, Chook YM, Taylor JP, Shorter J, Nuclear-Import Receptors Reverse Aberrant Phase Transitions of RNA-Binding Proteins with Prion-like Domains, Cell, 173 (2018) 677–692.e620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Jawerth L, Fischer-Friedrich E, Saha S, Wang J, Franzmann T, Zhang X, Sachweh J, Ruer M, Ijavi M, Saha S, Mahamid J, Hyman AA, Julicher F, Protein condensates as aging Maxwell fluids, Science, 370 (2020) 1317–1323. [DOI] [PubMed] [Google Scholar]
  • [54].Chuang E, Hori AM, Hesketh CD, Shorter J, Amyloid assembly and disassembly, J Cell Sci, 131 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Mathieu C, Pappu RV, Taylor JP, Beyond aggregation: Pathological phase transitions in neurodegenerative disease, Science, 370 (2020) 56–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Gitler AD, Dhillon P, Shorter J, Neurodegenerative disease: models, mechanisms, and a new hope, Dis Model Mech, 10 (2017) 499–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Shorter J, Designer protein disaggregases to counter neurodegenerative disease, Curr Opin Genet Dev, 44 (2017) 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Le Guerroue F, Youle RJ, Ubiquitin signaling in neurodegenerative diseases: an autophagy and proteasome perspective, Cell Death Differ, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Shorter J, Hsp104: a weapon to combat diverse neurodegenerative disorders, Neurosignals, 16 (2008) 63–74. [DOI] [PubMed] [Google Scholar]
  • [60].Uhlmann RE, Rother C, Rasmussen J, Schelle J, Bergmann C, Ullrich Gavilanes EM, Fritschi SK, Buehler A, Baumann F, Skodras A, Al-Shaana R, Beschorner N, Ye L, Kaeser SA, Obermuller U, Christensen S, Kartberg F, Stavenhagen JB, Rahfeld JU, Cynis H, Qian F, Weinreb PH, Bussiere T, Walker LC, Staufenbiel M, Jucker M, Acute targeting of pre-amyloid seeds in transgenic mice reduces Alzheimer-like pathology later in life, Nat Neurosci, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Guo L, Fare CM, Shorter J, Therapeutic Dissolution of Aberrant Phases by Nuclear-Import Receptors, Trends in cell biology, 29 (2019) 308–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Brown RH, Al-Chalabi A, Amyotrophic Lateral Sclerosis, N Engl J Med, 377 (2017) 162–172. [DOI] [PubMed] [Google Scholar]
  • [63].Taylor JP, Brown RH Jr., Cleveland DW, Decoding ALS: from genes to mechanism, Nature, 539 (2016) 197–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Kim G, Gautier O, Tassoni-Tsuchida E, Ma XR, Gitler AD, ALS Genetics: Gains, Losses, and Implications for Future Therapies, Neuron, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Lee S, Huang EJ, Modeling ALS and FTD with iPSC-derived neurons, Brain Res, 1656 (2017) 88–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].van Es MA, Hardiman O, Chio A, Al-Chalabi A, Pasterkamp RJ, Veldink JH, van den Berg LH, Amyotrophic lateral sclerosis, Lancet, 390 (2017) 2084–2098. [DOI] [PubMed] [Google Scholar]
  • [67].Harrison AF, Shorter J, RNA-binding proteins with prion-like domains in health and disease, Biochem J, 474 (2017) 1417–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Cook CN, Wu Y, Odeh HM, Gendron TF, Jansen-West K, Del Rosso G, Yue M, Jiang P, Gomes E, Tong J, Daughrity LM, Avendano NM, Castanedes-Casey M, Shao W, Oskarsson B, Tomassy GS, McCampbell A, Rigo F, Dickson DW, Shorter J, Zhang YJ, Petrucelli L, C9orf72 poly(GR) aggregation induces TDP-43 proteinopathy, Sci Transl Med, 12 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Odeh HM, Shorter J, Arginine-rich dipeptide-repeat proteins as phase disruptors in C9-ALS/FTD, Emerg Top Life Sci, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Ling SC, Polymenidou M, Cleveland DW, Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis, Neuron, 79 (2013) 416–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Nedelsky NB, Taylor JP, Bridging biophysics and neurology: aberrant phase transitions in neurodegenerative disease, Nat Rev Neurol, 15 (2019) 272–286. [DOI] [PubMed] [Google Scholar]
  • [72].Kedersha N, Stoecklin G, Ayodele M, Yacono P, Lykke-Andersen J, Fritzler MJ, Scheuner D, Kaufman RJ, Golan DE, Anderson P, Stress granules and processing bodies are dynamically linked sites of mRNP remodeling, J Cell Biol, 169 (2005) 871–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Cushman M, Johnson BS, King OD, Gitler AD, Shorter J, Prion-like disorders: blurring the divide between transmissibility and infectivity, J Cell Sci, 123 (2010) 1191–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].King OD, Gitler AD, Shorter J, The tip of the iceberg: RNA-binding proteins with prion-like domains in neurodegenerative disease, Brain Res, 1462 (2012) 61–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].March ZM, King OD, Shorter J, Prion-like domains as epigenetic regulators, scaffolds for subcellular organization, and drivers of neurodegenerative disease, Brain Res, 1647 (2016) 9–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Monahan Z, Ryan VH, Janke AM, Burke KA, Rhoads SN, Zerze GH, O’Meally R, Dignon GL, Conicella AE, Zheng W, Best RB, Cole RN, Mittal J, Shewmaker F, Fawzi NL, Phosphorylation of the FUS low-complexity domain disrupts phase separation, aggregation, and toxicity, Embo j, 36 (2017) 2951–2967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Owen I, Rhoads S, Yee D, Wyne H, Gery K, Hannula I, Sundrum M, Shewmaker F, The prion-like domain of Fused in Sarcoma is phosphorylated by multiple kinases affecting liquid- and solid-phase transitions, Mol Biol Cell, 31 (2020) 2522–2536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM, Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis, Science, 314 (2006) 130–133. [DOI] [PubMed] [Google Scholar]
  • [79].Neumann M, Kwong LK, Lee EB, Kremmer E, Flatley A, Xu Y, Forman MS, Troost D, Kretzschmar HA, Trojanowski JQ, Lee VM, Phosphorylation of S409/410 of TDP-43 is a consistent feature in all sporadic and familial forms of TDP-43 proteinopathies, Acta Neuropathol, 117 (2009) 137–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, Ackerley S, Durnall JC, Williams KL, Buratti E, Baralle F, de Belleroche J, Mitchell JD, Leigh PN, Al-Chalabi A, Miller CC, Nicholson G, Shaw CE, TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis, Science, 319 (2008) 1668–1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Johnson BS, Snead D, Lee JJ, McCaffery JM, Shorter J, Gitler AD, TDP-43 is intrinsically aggregation-prone, and amyotrophic lateral sclerosis-linked mutations accelerate aggregation and increase toxicity, J Biol Chem, 284 (2009) 20329–20339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Wang A, Conicella AE, Schmidt HB, Martin EW, Rhoads SN, Reeb AN, Nourse A, Ramirez Montero D, Ryan VH, Rohatgi R, Shewmaker F, Naik MT, Mittag T, Ayala YM, Fawzi NL, A single N-terminal phosphomimic disrupts TDP-43 polymerization, phase separation, and RNA splicing, Embo j, 37 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Bajc Česnik A, Motaln H, Rogelj B, The Impact of ALS-Associated Genes hnRNPA1, MATR3, VCP and UBQLN2 on the Severity of TDP-43 Aggregation, Cells, 9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Conicella AE, Dignon GL, Zerze GH, Schmidt HB, D’Ordine AM, Kim YC, Rohatgi R, Ayala YM, Mittal J, Fawzi NL, TDP-43 alpha-helical structure tunes liquid-liquid phase separation and function, Proc Natl Acad Sci U S A, 117 (2020) 5883–5894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Schmidt HB, Rohatgi R, In vivo formation of vacuolated multi-phase compartments lacking membranes, Cell reports, 16 (2016) 1228–1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Schmidt HB, Barreau A, Rohatgi R, Phase separation-deficient TDP43 remains functional in splicing, Nat Commun, 10 (2019) 4890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Shorter J, Taylor JP, Disease mutations in the prion-like domains of hnRNPA1 and hnRNPA2/B1 introduce potent steric zippers that drive excess RNP granule assembly, Rare Dis, 1 (2013) e25200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Taylor JP, Multisystem proteinopathy: intersecting genetics in muscle, bone, and brain degeneration, Neurology, 85 (2015) 658–660. [DOI] [PubMed] [Google Scholar]
  • [89].Lin Y, Protter DS, Rosen MK, Parker R, Formation and Maturation of Phase-Separated Liquid Droplets by RNA-Binding Proteins, Mol Cell, 60 (2015) 208–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Gilks N, Kedersha N, Ayodele M, Shen L, Stoecklin G, Dember LM, Anderson P, Stress granule assembly is mediated by prion-like aggregation of TIA-1, Mol Biol Cell, 15 (2004) 5383–5398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Mackenzie IR, Nicholson AM, Sarkar M, Messing J, Purice MD, Pottier C, Annu K, Baker M, Perkerson RB, Kurti A, Matchett BJ, Mittag T, Temirov J, Hsiung GR, Krieger C, Murray ME, Kato M, Fryer JD, Petrucelli L, Zinman L, Weintraub S, Mesulam M, Keith J, Zivkovic SA, Hirsch-Reinshagen V, Roos RP, Zuchner S, Graff-Radford NR, Petersen RC, Caselli RJ, Wszolek ZK, Finger E, Lippa C, Lacomis D, Stewart H, Dickson DW, Kim HJ, Rogaeva E, Bigio E, Boylan KB, Taylor JP, Rademakers R, TIA1 Mutations in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Promote Phase Separation and Alter Stress Granule Dynamics, Neuron, 95 (2017) 808–816.e809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, Kouri N, Wojtas A, Sengdy P, Hsiung GY, Karydas A, Seeley WW, Josephs KA, Coppola G, Geschwind DH, Wszolek ZK, Feldman H, Knopman DS, Petersen RC, Miller BL, Dickson DW, Boylan KB, Graff-Radford NR, Rademakers R, Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS, Neuron, 72 (2011) 245–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Renton AE, Majounie E, Waite A, Simón-Sánchez J, Rollinson S, Gibbs JR, Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L, Kalimo H, Paetau A, Abramzon Y, Remes AM, Kaganovich A, Scholz SW, Duckworth J, Ding J, Harmer DW, Hernandez DG, Johnson JO, Mok K, Ryten M, Trabzuni D, Guerreiro RJ, Orrell RW, Neal J, Murray A, Pearson J, Jansen IE, Sondervan D, Seelaar H, Blake D, Young K, Halliwell N, Callister JB, Toulson G, Richardson A, Gerhard A, Snowden J, Mann D, Neary D, Nalls MA, Peuralinna T, Jansson L, Isoviita VM, Kaivorinne AL, Hölttä-Vuori M, Ikonen E, Sulkava R, Benatar M, Wuu J, Chiò A, Restagno G, Borghero G, Sabatelli M, Heckerman D, Rogaeva E, Zinman L, Rothstein JD, Sendtner M, Drepper C, Eichler EE, Alkan C, Abdullaev Z, Pack SD, Dutra A, Pak E, Hardy J, Singleton A, Williams NM, Heutink P, Pickering-Brown S, Morris HR, Tienari PJ, Traynor BJ, A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD, Neuron, 72 (2011) 257–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Mori K, Weng SM, Arzberger T, May S, Rentzsch K, Kremmer E, Schmid B, Kretzschmar HA, Cruts M, Van Broeckhoven C, Haass C, Edbauer D, The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS, Science, 339 (2013) 1335–1338. [DOI] [PubMed] [Google Scholar]
  • [95].Zu T, Liu Y, Bañez-Coronel M, Reid T, Pletnikova O, Lewis J, Miller TM, Harms MB, Falchook AE, Subramony SH, Ostrow LW, Rothstein JD, Troncoso JC, Ranum LP, RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia, Proc Natl Acad Sci U S A, 110 (2013) E4968–4977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].McEachin ZT, Gendron TF, Raj N, Garcia-Murias M, Banerjee A, Purcell RH, Ward PJ, Todd TW, Merritt-Garza ME, Jansen-West K, Hales CM, Garcia-Sobrino T, Quintans B, Holler CJ, Taylor G, San Millan B, Teijeira S, Yamashita T, Ohkubo R, Boulis NM, Xu C, Wen Z, Streichenberger N, Neuro CEBNN, Fogel BL, Kukar T, Abe K, Dickson DW, Arias M, Glass JD, Jiang J, Tansey MG, Sobrido MJ, Petrucelli L, Rossoll W, Bassell GJ, Chimeric Peptide Species Contribute to Divergent Dipeptide Repeat Pathology in c9ALS/FTD and SCA36, Neuron, 107 (2020) 292–305 e296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Saberi S, Stauffer JE, Jiang J, Garcia SD, Taylor AE, Schulte D, Ohkubo T, Schloffman CL, Maldonado M, Baughn M, Rodriguez MJ, Pizzo D, Cleveland D, Ravits J, Sense-encoded poly-GR dipeptide repeat proteins correlate to neurodegeneration and uniquely co-localize with TDP-43 in dendrites of repeat-expanded C9orf72 amyotrophic lateral sclerosis, Acta Neuropathol, 135 (2018) 459–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Lee KH, Zhang P, Kim HJ, Mitrea DM, Sarkar M, Freibaum BD, Cika J, Coughlin M, Messing J, Molliex A, Maxwell BA, Kim NC, Temirov J, Moore J, Kolaitis RM, Shaw TI, Bai B, Peng J, Kriwacki RW, Taylor JP, C9orf72 Dipeptide Repeats Impair the Assembly, Dynamics, and Function of Membrane-Less Organelles, Cell, 167 (2016) 774–788.e717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Boeynaems S, Holehouse AS, Weinhardt V, Kovacs D, Van Lindt J, Larabell C, Van Den Bosch L, Das R, Tompa PS, Pappu RV, Gitler AD, Spontaneous driving forces give rise to protein-RNA condensates with coexisting phases and complex material properties, Proc Natl Acad Sci U S A, 116 (2019) 7889–7898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Boeynaems S, Bogaert E, Kovacs D, Konijnenberg A, Timmerman E, Volkov A, Guharoy M, De Decker M, Jaspers T, Ryan VH, Janke AM, Baatsen P, Vercruysse T, Kolaitis RM, Daelemans D, Taylor JP, Kedersha N, Anderson P, Impens F, Sobott F, Schymkowitz J, Rousseau F, Fawzi NL, Robberecht W, Van Damme P, Tompa P, Van Den Bosch L, Phase Separation of C9orf72 Dipeptide Repeats Perturbs Stress Granule Dynamics, Mol Cell, 65 (2017) 1044–1055 e1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Hutten S, Usluer S, Bourgeois B, Simonetti F, Odeh HM, Fare CM, Czuppa M, Hruska-Plochan M, Hofweber M, Polymenidou M, Shorter J, Edbauer D, Madl T, Dormann D, Nuclear import receptors directly bind to arginine-rich dipeptide repeat proteins and suppress their pathological interactions., Cell Rep, 33 (2020) 108538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Elden AC, Kim HJ, Hart MP, Chen-Plotkin AS, Johnson BS, Fang X, Armakola M, Geser F, Greene R, Lu MM, Padmanabhan A, Clay-Falcone D, McCluskey L, Elman L, Juhr D, Gruber PJ, Rüb U, Auburger G, Trojanowski JQ, Lee VM, Van Deerlin VM, Bonini NM, Gitler AD, Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS, Nature, 466 (2010) 1069–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Lee T, Li YR, Chesi A, Hart MP, Ramos D, Jethava N, Hosangadi D, Epstein J, Hodges B, Bonini NM, Gitler AD, Evaluating the prevalence of polyglutamine repeat expansions in amyotrophic lateral sclerosis, Neurology, 76 (2011) 2062–2065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Lee T, Li YR, Ingre C, Weber M, Grehl T, Gredal O, de Carvalho M, Meyer T, Tysnes OB, Auburger G, Gispert S, Bonini NM, Andersen PM, Gitler AD, Ataxin-2 intermediate-length polyglutamine expansions in European ALS patients, Hum Mol Genet, 20 (2011) 1697–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Yu Z, Zhu Y, Chen-Plotkin AS, Clay-Falcone D, McCluskey L, Elman L, Kalb RG, Trojanowski JQ, Lee VM, Van Deerlin VM, Gitler AD, Bonini NM, PolyQ repeat expansions in ATXN2 associated with ALS are CAA interrupted repeats, PLoS One, 6 (2011) e17951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Imbert G, Saudou F, Yvert G, Devys D, Trottier Y, Garnier JM, Weber C, Mandel JL, Cancel G, Abbas N, Durr A, Didierjean O, Stevanin G, Agid Y, Brice A, Cloning of the gene for spinocerebellar ataxia 2 reveals a locus with high sensitivity to expanded CAG/glutamine repeats, Nat Genet, 14 (1996) 285–291. [DOI] [PubMed] [Google Scholar]
  • [107].Pulst SM, Nechiporuk A, Nechiporuk T, Gispert S, Chen XN, Lopes-Cendes I, Pearlman S, Starkman S, Orozco-Diaz G, Lunkes A, DeJong P, Rouleau GA, Auburger G, Korenberg JR, Figueroa C, Sahba S, Moderate expansion of a normally biallelic trinucleotide repeat in spinocerebellar ataxia type 2, Nat Genet, 14 (1996) 269–276. [DOI] [PubMed] [Google Scholar]
  • [108].Sanpei K, Takano H, Igarashi S, Sato T, Oyake M, Sasaki H, Wakisaka A, Tashiro K, Ishida Y, Ikeuchi T, Koide R, Saito M, Sato A, Tanaka T, Hanyu S, Takiyama Y, Nishizawa M, Shimizu N, Nomura Y, Segawa M, Iwabuchi K, Eguchi I, Tanaka H, Takahashi H, Tsuji S, Identification of the spinocerebellar ataxia type 2 gene using a direct identification of repeat expansion and cloning technique, DIRECT, Nat Genet, 14 (1996) 277–284. [DOI] [PubMed] [Google Scholar]
  • [109].Malinovska L, Kroschwald S, Alberti S, Protein disorder, prion propensities, and self-organizing macromolecular collectives, Biochim Biophys Acta, 1834 (2013) 918–931. [DOI] [PubMed] [Google Scholar]
  • [110].Becker LA, Huang B, Bieri G, Ma R, Knowles DA, Jafar-Nejad P, Messing J, Kim HJ, Soriano A, Auburger G, Pulst SM, Taylor JP, Rigo F, Gitler AD, Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice, Nature, 544 (2017) 367–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Zhang C, Saunders AJ, An emerging role for Ubiquilin 1 in regulating protein quality control system and in disease pathogenesis, Discov Med, 8 (2009) 18–22. [PMC free article] [PubMed] [Google Scholar]
  • [112].Deng HX, Chen W, Hong ST, Boycott KM, Gorrie GH, Siddique N, Yang Y, Fecto F, Shi Y, Zhai H, Jiang H, Hirano M, Rampersaud E, Jansen GH, Donkervoort S, Bigio EH, Brooks BR, Ajroud K, Sufit RL, Haines JL, Mugnaini E, Pericak-Vance MA, Siddique T, Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia, Nature, 477 (2011) 211–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Higgins N, Lin B, Monteiro MJ, Lou Gehrig’s Disease (ALS): UBQLN2 Mutations Strike Out of Phase, Structure, 27 (2019) 879–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Selkoe DJ, Hardy J, The amyloid hypothesis of Alzheimer’s disease at 25 years, EMBO Mol Med, 8 (2016) 595–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Gibbons GS, Kim SJ, Wu Q, Riddle DM, Leight SN, Changolkar L, Xu H, Meymand ES, O’Reilly M, Zhang B, Brunden KR, Trojanowski JQ, Lee VMY, Conformation-selective tau monoclonal antibodies inhibit tau pathology in primary neurons and a mouse model of Alzheimer’s disease, Mol Neurodegener, 15 (2020) 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Vanderweyde T, Yu H, Varnum M, Liu-Yesucevitz L, Citro A, Ikezu T, Duff K, Wolozin B, Contrasting pathology of the stress granule proteins TIA-1 and G3BP in tauopathies, J Neurosci, 32 (2012) 8270–8283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Diner I, Hales CM, Bishof I, Rabenold L, Duong DM, Yi H, Laur O, Gearing M, Troncoso J, Thambisetty M, Lah JJ, Levey AI, Seyfried NT, Aggregation properties of the small nuclear ribonucleoprotein U1–70K in Alzheimer disease, J Biol Chem, 289 (2014) 35296–35313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Ballatore C, Lee VM, Trojanowski JQ, Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders, Nat Rev Neurosci, 8 (2007) 663–672. [DOI] [PubMed] [Google Scholar]
  • [119].Drubin DG, Kirschner MW, Tau protein function in living cells, J Cell Biol, 103 (1986) 2739–2746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Lee VM, Balin BJ, Otvos L Jr., Trojanowski JQ, A68: a major subunit of paired helical filaments and derivatized forms of normal Tau, Science, 251 (1991) 675–678. [DOI] [PubMed] [Google Scholar]
  • [121].Mucke L, Neuroscience: Alzheimer’s disease, Nature, 461 (2009) 895–897. [DOI] [PubMed] [Google Scholar]
  • [122].Williams DR, Tauopathies: classification and clinical update on neurodegenerative diseases associated with microtubule-associated protein tau, Intern Med J, 36 (2006) 652–660. [DOI] [PubMed] [Google Scholar]
  • [123].Kovacs GG, Tauopathies, Handb Clin Neurol, 145 (2017) 355–368. [DOI] [PubMed] [Google Scholar]
  • [124].Olszewska DA, Lonergan R, Fallon EM, Lynch T, Genetics of Frontotemporal Dementia, Curr Neurol Neurosci Rep, 16 (2016) 107. [DOI] [PubMed] [Google Scholar]
  • [125].Andorfer C, Kress Y, Espinoza M, de Silva R, Tucker KL, Barde YA, Duff K, Davies P, Hyperphosphorylation and aggregation of tau in mice expressing normal human tau isoforms, J Neurochem, 86 (2003) 582–590. [DOI] [PubMed] [Google Scholar]
  • [126].Xia Y, Prokop S, Gorion KM, Kim JD, Sorrentino ZA, Bell BM, Manaois AN, Chakrabarty P, Davies P, Giasson BI, Tau Ser208 phosphorylation promotes aggregation and reveals neuropathologic diversity in Alzheimer’s disease and other tauopathies, Acta Neuropathol Commun, 8 (2020) 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Despres C, Byrne C, Qi H, Cantrelle FX, Huvent I, Chambraud B, Baulieu EE, Jacquot Y, Landrieu I, Lippens G, Smet-Nocca C, Identification of the Tau phosphorylation pattern that drives its aggregation, Proc Natl Acad Sci U S A, 114 (2017) 9080–9085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Young ZT, Mok SA, Gestwicki JE, Therapeutic Strategies for Restoring Tau Homeostasis, Cold Spring Harb Perspect Med, 8 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Bulic B, Pickhardt M, Mandelkow EM, Mandelkow E, Tau protein and tau aggregation inhibitors, Neuropharmacology, 59 (2010) 276–289. [DOI] [PubMed] [Google Scholar]
  • [130].Hernandez-Vega A, Braun M, Scharrel L, Jahnel M, Wegmann S, Hyman BT, Alberti S, Diez S, Hyman AA, Local Nucleation of Microtubule Bundles through Tubulin Concentration into a Condensed Tau Phase, Cell Rep, 20 (2017) 2304–2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Ambadipudi S, Biernat J, Riedel D, Mandelkow E, Zweckstetter M, Liquid-liquid phase separation of the microtubule-binding repeats of the Alzheimer-related protein Tau, Nat Commun, 8 (2017) 275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Kanaan NM, Hamel C, Grabinski T, Combs B, Liquid-liquid phase separation induces pathogenic tau conformations in vitro, Nat Commun, 11 (2020) 2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Zhang X, Lin Y, Eschmann NA, Zhou H, Rauch JN, Hernandez I, Guzman E, Kosik KS, Han S, RNA stores tau reversibly in complex coacervates, PLoS Biol, 15 (2017) e2002183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Ferreon JC, Jain A, Choi KJ, Tsoi PS, MacKenzie KR, Jung SY, Ferreon AC, Acetylation Disfavors Tau Phase Separation, Int J Mol Sci, 19 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Bai B, Hales CM, Chen PC, Gozal Y, Dammer EB, Fritz JJ, Wang X, Xia Q, Duong DM, Street C, Cantero G, Cheng D, Jones DR, Wu Z, Li Y, Diner I, Heilman CJ, Rees HD, Wu H, Lin L, Szulwach KE, Gearing M, Mufson EJ, Bennett DA, Montine TJ, Seyfried NT, Wingo TS, Sun YE, Jin P, Hanfelt J, Willcock DM, Levey A, Lah JJ, Peng J, U1 small nuclear ribonucleoprotein complex and RNA splicing alterations in Alzheimer’s disease, Proc Natl Acad Sci U S A, 110 (2013) 16562–16567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Xue S, Gong R, He F, Li Y, Wang Y, Tan T, Luo SZ, Low-complexity domain of U1–70K modulates phase separation and aggregation through distinctive basic-acidic motifs, Sci Adv, 5 (2019) eaax5349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Bishof I, Dammer EB, Duong DM, Kundinger SR, Gearing M, Lah JJ, Levey AI, Seyfried NT, RNA-binding proteins with basic-acidic dipeptide (BAD) domains self-assemble and aggregate in Alzheimer’s disease, J Biol Chem, 293 (2018) 11047–11066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Beitz JM, Parkinson’s disease: a review, Front Biosci (Schol Ed), 6 (2014) 65–74. [DOI] [PubMed] [Google Scholar]
  • [139].Spillantini MG, Crowther RA, Jakes R, Cairns NJ, Lantos PL, Goedert M, Filamentous alpha-synuclein inclusions link multiple system atrophy with Parkinson’s disease and dementia with Lewy bodies, Neurosci Lett, 251 (1998) 205–208. [DOI] [PubMed] [Google Scholar]
  • [140].Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M, alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies, Proc Natl Acad Sci U S A, 95 (1998) 6469–6473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M, Alpha-synuclein in Lewy bodies, Nature, 388 (1997) 839–840. [DOI] [PubMed] [Google Scholar]
  • [142].Fanning S, Selkoe D, Dettmer U, Parkinson’s disease: proteinopathy or lipidopathy?, NPJ Parkinsons Dis, 6 (2020) 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, Lee VM, Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice, Science, 338 (2012) 949–953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, Meaney DF, Trojanowski JQ, Lee VM, Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death, Neuron, 72 (2011) 57–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Froula JM, Castellana-Cruz M, Anabtawi NM, Camino JD, Chen SW, Thrasher DR, Freire J, Yazdi AA, Fleming S, Dobson CM, Kumita JR, Cremades N, Volpicelli-Daley LA, Defining alpha-synuclein species responsible for Parkinson’s disease phenotypes in mice, J Biol Chem, 294 (2019) 10392–10406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Mahul-Mellier AL, Burtscher J, Maharjan N, Weerens L, Croisier M, Kuttler F, Leleu M, Knott GW, Lashuel HA, The process of Lewy body formation, rather than simply alpha-synuclein fibrillization, is one of the major drivers of neurodegeneration, Proc Natl Acad Sci U S A, 117 (2020) 4971–4982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Fares MB, Jagannath S, Lashuel HA, Reverse engineering Lewy bodies: how far have we come and how far can we go?, Nat Rev Neurosci, (2021). [DOI] [PubMed] [Google Scholar]
  • [148].Lo Bianco C, Shorter J, Regulier E, Lashuel H, Iwatsubo T, Lindquist S, Aebischer P, Hsp104 antagonizes alpha-synuclein aggregation and reduces dopaminergic degeneration in a rat model of Parkinson disease, J Clin Invest, 118 (2008) 3087–3097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Zhu G, Harischandra DS, Ghaisas S, Zhang P, Prall W, Huang L, Maghames C, Guo L, Luna E, Mack KL, Torrente MP, Luk KC, Shorter J, Yang X, TRIM11 Prevents and Reverses Protein Aggregation and Rescues a Mouse Model of Parkinson’s Disease, Cell Rep, 33 (2020) 108418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Lashuel HA, Overk CR, Oueslati A, Masliah E, The many faces of α-synuclein: from structure and toxicity to therapeutic target, Nature reviews. Neuroscience, 14 (2013) 38–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Crowther RA, Jakes R, Spillantini MG, Goedert M, Synthetic filaments assembled from C-terminally truncated alpha-synuclein, FEBS letters, 436 (1998) 309–312. [DOI] [PubMed] [Google Scholar]
  • [152].Schweighauser M, Shi Y, Tarutani A, Kametani F, Murzin AG, Ghetti B, Matsubara T, Tomita T, Ando T, Hasegawa K, Murayama S, Yoshida M, Hasegawa M, Scheres SHW, Goedert M, Structures of α-synuclein filaments from multiple system atrophy, Nature, 585 (2020) 464–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Yasuda T, Nakata Y, Mochizuki H, α-Synuclein and neuronal cell death, Molecular neurobiology, 47 (2013) 466–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Hardenberg MC, Sinnige T, Casford S, Dada S, Poudel C, Robinson EA, Fuxreiter M, Kaminksi C, Kaminski-Schierle GS, Nollen EAA, Dobson CM, Vendruscolo M, Observation of an alpha-synuclein liquid droplet state and its maturation into Lewy body-like assemblies, J Mol Cell Biol, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Shahmoradian SH, Lewis AJ, Genoud C, Hench J, Moors TE, Navarro PP, Castano-Diez D, Schweighauser G, Graff-Meyer A, Goldie KN, Sutterlin R, Huisman E, Ingrassia A, Gier Y, Rozemuller AJM, Wang J, Paepe A, Erny J, Staempfli A, Hoernschemeyer J, Grosseruschkamp F, Niedieker D, El-Mashtoly SF, Quadri M, Van IWFJ, Bonifati V, Gerwert K, Bohrmann B, Frank S, Britschgi M, Stahlberg H, Van de Berg WDJ, Lauer ME, Lewy pathology in Parkinson’s disease consists of crowded organelles and lipid membranes, Nat Neurosci, 22 (2019) 1099–1109. [DOI] [PubMed] [Google Scholar]
  • [156].Araki K, Yagi N, Aoyama K, Choong CJ, Hayakawa H, Fujimura H, Nagai Y, Goto Y, Mochizuki H, Parkinson’s disease is a type of amyloidosis featuring accumulation of amyloid fibrils of alpha-synuclein, Proc Natl Acad Sci U S A, 116 (2019) 17963–17969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Lashuel HA, Do Lewy bodies contain alpha-synuclein fibrils? and Does it matter? A brief history and critical analysis of recent reports, Neurobiol Dis, 141 (2020) 104876. [DOI] [PubMed] [Google Scholar]
  • [158].Hatters DM, Protein misfolding inside cells: the case of huntingtin and Huntington’s disease, IUBMB Life, 60 (2008) 724–728. [DOI] [PubMed] [Google Scholar]
  • [159].Myers RH, Huntington’s disease genetics, NeuroRx, 1 (2004) 255–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Squitieri F, Cannella M, Simonelli M, CAG mutation effect on rate of progression in Huntington’s disease, Neurol Sci, 23 Suppl 2 (2002) S107–108. [DOI] [PubMed] [Google Scholar]
  • [161].Dewan R, Chia R, Ding J, Hickman RA, Stein TD, Abramzon Y, Ahmed S, Sabir MS, Portley MK, Tucci A, Ibáñez K, Shankaracharya FNU, Keagle P, Rossi G, Caroppo P, Tagliavini F, Waldo ML, Johansson PM, Nilsson CF, Rowe JB, Benussi L, Binetti G, Ghidoni R, Jabbari E, Viollet C, Glass JD, Singleton AB, Silani V, Ross OA, Ryten M, Torkamani A, Tanaka T, Ferrucci L, Resnick SM, Pickering-Brown S, Brady CB, Kowal N, Hardy JA, Van Deerlin V, Vonsattel JP, Harms MB, Morris HR, Ferrari R, Landers JE, Chiò A, Gibbs JR, Dalgard CL, Scholz SW, Traynor BJ, Pathogenic Huntingtin Repeat Expansions in Patients with Frontotemporal Dementia and Amyotrophic Lateral Sclerosis, Neuron, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Balch WE, Morimoto RI, Dillin A, Kelly JW, Adapting proteostasis for disease intervention, Science, 319 (2008) 916–919. [DOI] [PubMed] [Google Scholar]
  • [163].Valastyan JS, Lindquist S, Mechanisms of protein-folding diseases at a glance, Dis Model Mech, 7 (2014) 9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Klaips CL, Jayaraj GG, Hartl FU, Pathways of cellular proteostasis in aging and disease, J Cell Biol, 217 (2018) 51–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Morimoto RI, Stress, aging, and neurodegenerative disease, N Engl J Med, 355 (2006) 2254–2255. [DOI] [PubMed] [Google Scholar]
  • [166].Shorter J, Southworth DR, Spiraling in Control: Structures and Mechanisms of the Hsp104 Disaggregase, Cold Spring Harb Perspect Biol, 11 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Torrente MP, Shorter J, The metazoan protein disaggregase and amyloid depolymerase system: Hsp110, Hsp70, Hsp40, and small heat shock proteins, Prion, 7 (2013) 457–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Cupo RR, Shorter J, Skd3 (human ClpB) is a potent mitochondrial protein disaggregase that is inactivated by 3-methylglutaconic aciduria-linked mutations, Elife, 9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Smith HL, Li W, Cheetham ME, Molecular chaperones and neuronal proteostasis, Semin Cell Dev Biol, 40 (2015) 142–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Farhan SMK, Howrigan DP, Abbott LE, Klim JR, Topp SD, Byrnes AE, Churchhouse C, Phatnani H, Smith BN, Rampersaud E, Wu G, Wuu J, Shatunov A, Iacoangeli A, Al Khleifat A, Mordes DA, Ghosh S, Consortium A, Consortium F, Project Min EC, Consortium CR, Eggan K, Rademakers R, McCauley JL, Schule R, Zuchner S, Benatar M, Taylor JP, Nalls M, Gotkine M, Shaw PJ, Morrison KE, Al-Chalabi A, Traynor B, Shaw CE, Goldstein DB, Harms MB, Daly MJ, Neale BM, Exome sequencing in amyotrophic lateral sclerosis implicates a novel gene, DNAJC7, encoding a heat-shock protein, Nat Neurosci, 22 (2019) 1966–1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Roosen DA, Blauwendraat C, Cookson MR, Lewis PA, DNAJC proteins and pathways to parkinsonism, FEBS J, 286 (2019) 3080–3094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Wu YR, Wang CK, Chen CM, Hsu Y, Lin SJ, Lin YY, Fung HC, Chang KH, Lee-Chen GJ, Analysis of heat-shock protein 70 gene polymorphisms and the risk of Parkinson’s disease, Hum Genet, 114 (2004) 236–241. [DOI] [PubMed] [Google Scholar]
  • [173].Lindberg I, Shorter J, Wiseman RL, Chiti F, Dickey CA, McLean PJ, Chaperones in Neurodegeneration, The Journal of neuroscience : the official journal of the Society for Neuroscience, 35 (2015) 13853–13859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Mack KL, Shorter J, Engineering and Evolution of Molecular Chaperones and Protein Disaggregases with Enhanced Activity, Frontiers in molecular biosciences, 3 (2016) 8–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Bonini NM, Chaperoning brain degeneration, Proc Natl Acad Sci U S A, 99 Suppl 4 (2002) 16407–16411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].March ZM, Mack KL, Shorter J, AAA+ Protein-Based Technologies to Counter Neurodegenerative Disease, Biophys J, 116 (2019) 1380–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Cushman-Nick M, Bonini NM, Shorter J, Hsp104 suppresses polyglutamine-induced degeneration post onset in a drosophila MJD/SCA3 model, PLoS Genet, 9 (2013) e1003781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Auluck PK, Chan HY, Trojanowski JQ, Lee VM, Bonini NM, Chaperone suppression of alpha-synuclein toxicity in a Drosophila model for Parkinson’s disease, Science, 295 (2002) 865–868. [DOI] [PubMed] [Google Scholar]
  • [179].Warrick JM, Chan HY, Gray-Board GL, Chai Y, Paulson HL, Bonini NM, Suppression of polyglutamine-mediated neurodegeneration in Drosophila by the molecular chaperone HSP70, Nat Genet, 23 (1999) 425–428. [DOI] [PubMed] [Google Scholar]
  • [180].Nagy M, Fenton WA, Li D, Furtak K, Horwich AL, Extended survival of misfolded G85R SOD1-linked ALS mice by transgenic expression of chaperone Hsp110, Proc Natl Acad Sci U S A, 113 (2016) 5424–5428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Taguchi YV, Gorenberg EL, Nagy M, Thrasher D, Fenton WA, Volpicelli-Daley L, Horwich AL, Chandra SS, Hsp110 mitigates alpha-synuclein pathology in vivo, Proc Natl Acad Sci U S A, 116 (2019) 24310–24316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Shorter J, Engineering therapeutic protein disaggregases, Mol Biol Cell, 27 (2016) 1556–1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Jackrel ME, Shorter J, Engineering enhanced protein disaggregases for neurodegenerative disease, Prion, 9 (2015) 90–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Tariq A, Lin J, Jackrel ME, Hesketh CD, Carman PJ, Mack KL, Weitzman R, Gambogi C, Hernandez Murillo OA, Sweeny EA, Gurpinar E, Yokom AL, Gates SN, Yee K, Sudesh S, Stillman J, Rizo AN, Southworth DR, Shorter J, Mining Disaggregase Sequence Space to Safely Counter TDP-43, FUS, and alpha-Synuclein Proteotoxicity, Cell Rep, 28 (2019) 2080–2095.e2086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Tariq A, Lin J, Noll MM, Torrente MP, Mack KL, Murillo OH, Jackrel ME, Shorter J, Potentiating Hsp104 activity via phosphomimetic mutations in the middle domain, FEMS Yeast Res, 18 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Desantis ME, Shorter J, The elusive middle domain of Hsp104 and ClpB: location and function, Biochim Biophys Acta, 1823 (2012) 29–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Sweeny EA, Shorter J, Mechanistic and Structural Insights into the Prion-Disaggregase Activity of Hsp104, J Mol Biol, 428 (2016) 1870–1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Jackrel ME, DeSantis ME, Martinez BA, Castellano LM, Stewart RM, Caldwell KA, Caldwell GA, Shorter J, Potentiated Hsp104 variants antagonize diverse proteotoxic misfolding events, Cell, 156 (2014) 170–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Yasuda K, Clatterbuck-Soper SF, Jackrel ME, Shorter J, Mili S, FUS inclusions disrupt RNA localization by sequestering kinesin-1 and inhibiting microtubule detyrosination, J Cell Biol, 216 (2017) 1015–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].March ZM, Sweeney K, Kim H, Yan X, Castellano LM, Jackrel ME, Lin J, Chuang E, Gomes E, Willicott CW, Michalska K, Jedrzejczak RP, Joachimiak A, Caldwell KA, Caldwell GA, Shalem O, Shorter J, Therapeutic genetic variation revealed in diverse Hsp104 homologs, Elife, 9 (2020) 2020.2003.2025.008748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Mack KL, Kim H, Jackrel ME, Lin J, DeNizio JE, Yan X, Chuang E, Tariq A, Cupo RR, Castellano LM, Caldwell KA, Caldwell GA, Shorter J, Tuning Hsp104 specificity to selectively detoxify α-synuclein, bioRxiv, (2020) 2020.2004.2015.043935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Darwich NF, Phan JM, Kim B, Suh E, Papatriantafyllou JD, Changolkar L, Nguyen AT, O’Rourke CM, He Z, Porta S, Gibbons GS, Luk KC, Papageorgiou SG, Grossman M, Massimo L, Irwin DJ, McMillan CT, Nasrallah IM, Toro C, Aguirre GK, Van Deerlin VM, Lee EB, Autosomal dominant VCP hypomorph mutation impairs disaggregation of PHF-tau, Science, 370 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Wang B, Maxwell BA, Joo JH, Gwon Y, Messing J, Mishra A, Shaw TI, Ward AL, Quan H, Sakurada SM, Pruett-Miller SM, Bertorini T, Vogel P, Kim HJ, Peng J, Taylor JP, Kundu M, ULK1 and ULK2 Regulate Stress Granule Disassembly Through Phosphorylation and Activation of VCP/p97, Mol Cell, 74 (2019) 742–757 e748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Duennwald ML, Echeverria A, Shorter J, Small heat shock proteins potentiate amyloid dissolution by protein disaggregases from yeast and humans, PLoS Biol, 10 (2012) e1001346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Shorter J, The mammalian disaggregase machinery: Hsp110 synergizes with Hsp70 and Hsp40 to catalyze protein disaggregation and reactivation in a cell-free system, PLoS One, 6 (2011) e26319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Faust O, Abayev-Avraham M, Wentink AS, Maurer M, Nillegoda NB, London N, Bukau B, Rosenzweig R, HSP40 proteins use class-specific regulation to drive HSP70 functional diversity, Nature, 587 (2020) 489–494. [DOI] [PubMed] [Google Scholar]
  • [197].Gao X, Carroni M, Nussbaum-Krammer C, Mogk A, Nillegoda NB, Szlachcic A, Guilbride DL, Saibil HR, Mayer MP, Bukau B, Human Hsp70 Disaggregase Reverses Parkinson’s-Linked alpha-Synuclein Amyloid Fibrils, Mol Cell, 59 (2015) 781–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Nillegoda NB, Kirstein J, Szlachcic A, Berynskyy M, Stank A, Stengel F, Arnsburg K, Gao X, Scior A, Aebersold R, Guilbride DL, Wade RC, Morimoto RI, Mayer MP, Bukau B, Crucial HSP70 co-chaperone complex unlocks metazoan protein disaggregation, Nature, 524 (2015) 247–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Wentink AS, Nillegoda NB, Feufel J, Ubartaite G, Schneider CP, De Los Rios P, Hennig J, Barducci A, Bukau B, Molecular dissection of amyloid disaggregation by human HSP70, Nature, 587 (2020) 483–488. [DOI] [PubMed] [Google Scholar]
  • [200].Scior A, Buntru A, Arnsburg K, Ast A, Iburg M, Juenemann K, Pigazzini ML, Mlody B, Puchkov D, Priller J, Wanker EE, Prigione A, Kirstein J, Complete suppression of Htt fibrilization and disaggregation of Htt fibrils by a trimeric chaperone complex, EMBO J, 37 (2018) 282–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Yu H, Lu S, Gasior K, Singh D, Vazquez-Sanchez S, Tapia O, Toprani D, Beccari MS, Yates JR 3rd, Da Cruz S, Newby JM, Lafarga M, Gladfelter AS, Villa E, Cleveland DW, HSP70 chaperones RNA-free TDP-43 into anisotropic intranuclear liquid spherical shells, Science, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Aponte RA, Zimmermann S, Reinstein J, Directed Evolution of the DnaK Chaperone: Mutations in the Lid Domain Result in Enhanced Chaperone Activity, Journal of Molecular Biology, 399 (2010) 154–167. [DOI] [PubMed] [Google Scholar]
  • [203].Lindstedt PR, Aprile FA, Matos MJ, Perni M, Bertoldo JB, Bernardim B, Peter Q, Jimenez-Oses G, Knowles TPJ, Dobson CM, Corzana F, Vendruscolo M, Bernardes GJL, Enhancement of the Anti-Aggregation Activity of a Molecular Chaperone Using a Rationally Designed Post-Translational Modification, ACS Cent Sci, 5 (2019) 1417–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Aprile FA, Sormanni P, Vendruscolo M, A Rational Design Strategy for the Selective Activity Enhancement of a Molecular Chaperone toward a Target Substrate, Biochemistry, 54 (2015) 5103–5112. [DOI] [PubMed] [Google Scholar]
  • [205].Bongiovanni MN, Aprile FA, Sormanni P, Vendruscolo M, A Rationally Designed Hsp70 Variant Rescues the Aggregation-Associated Toxicity of Human IAPP in Cultured Pancreatic Islet beta-Cells, Int J Mol Sci, 19 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Guo L, Giasson BI, Glavis-Bloom A, Brewer MD, Shorter J, Gitler AD, Yang X, A cellular system that degrades misfolded proteins and protects against neurodegeneration, Mol Cell, 55 (2014) 15–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Zhang L, Afolabi LO, Wan X, Li Y, Chen L, Emerging Roles of Tripartite Motif-Containing Family Proteins (TRIMs) in Eliminating Misfolded Proteins, Front Cell Dev Biol, 8 (2020) 802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Dhar N, Arsiwala A, Murali S, Kane RS, “Trim”ming PolyQ proteins with engineered PML, Biotechnol Bioeng, 117 (2020) 362–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY, Gene therapy for neurological disorders: progress and prospects, Nat Rev Drug Discov, 17 (2018) 641–659. [DOI] [PubMed] [Google Scholar]
  • [210].Naldini L, Gene therapy returns to centre stage, Nature, 526 (2015) 351–360. [DOI] [PubMed] [Google Scholar]
  • [211].LeWitt PA, Rezai AR, Leehey MA, Ojemann SG, Flaherty AW, Eskandar EN, Kostyk SK, Thomas K, Sarkar A, Siddiqui MS, Tatter SB, Schwalb JM, Poston KL, Henderson JM, Kurlan RM, Richard IH, Van Meter L, Sapan CV, During MJ, Kaplitt MG, Feigin A, AAV2-GAD gene therapy for advanced Parkinson’s disease: a double-blind, sham-surgery controlled, randomised trial, Lancet Neurol, 10 (2011) 309–319. [DOI] [PubMed] [Google Scholar]
  • [212].Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW, Lowes L, Alfano L, Berry K, Church K, Kissel JT, Nagendran S, L’Italien J, Sproule DM, Wells C, Cardenas JA, Heitzer MD, Kaspar A, Corcoran S, Braun L, Likhite S, Miranda C, Meyer K, Foust KD, Burghes AHM, Kaspar BK, Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy, N Engl J Med, 377 (2017) 1713–1722. [DOI] [PubMed] [Google Scholar]
  • [213].Warren Olanow C, Bartus RT, Baumann TL, Factor S, Boulis N, Stacy M, Turner DA, Marks W, Larson P, Starr PA, Jankovic J, Simpson R, Watts R, Guthrie B, Poston K, Henderson JM, Stern M, Baltuch G, Goetz CG, Herzog C, Kordower JH, Alterman R, Lozano AM, Lang AE, Gene delivery of neurturin to putamen and substantia nigra in Parkinson disease: A double-blind, randomized, controlled trial, Ann Neurol, 78 (2015) 248–257. [DOI] [PubMed] [Google Scholar]
  • [214].Jackrel ME, Shorter J, Shock and awe: unleashing the heat shock response to treat Huntington disease, J Clin Invest, 121 (2011) 2972–2975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Auluck PK, Bonini NM, Pharmacological prevention of Parkinson disease in Drosophila, Nat Med, 8 (2002) 1185–1186. [DOI] [PubMed] [Google Scholar]
  • [216].Labbadia J, Cunliffe H, Weiss A, Katsyuba E, Sathasivam K, Seredenina T, Woodman B, Moussaoui S, Frentzel S, Luthi-Carter R, Paganetti P, Bates GP, Altered chromatin architecture underlies progressive impairment of the heat shock response in mouse models of Huntington disease, J Clin Invest, 121 (2011) 3306–3319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Sabath N, Levy-Adam F, Younis A, Rozales K, Meller A, Hadar S, Soueid-Baumgarten S, Shalgi R, Cellular proteostasis decline in human senescence, Proc Natl Acad Sci U S A, 117 (2020) 31902–31913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Chou A, Krukowski K, Jopson T, Zhu PJ, Costa-Mattioli M, Walter P, Rosi S, Inhibition of the integrated stress response reverses cognitive deficits after traumatic brain injury, Proc Natl Acad Sci U S A, 114 (2017) E6420–E6426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Krukowski K, Nolan A, Frias ES, Boone M, Ureta G, Grue K, Paladini MS, Elizarraras E, Delgado L, Bernales S, Walter P, Rosi S, Small molecule cognitive enhancer reverses age-related memory decline in mice, Elife, 9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Krukowski K, Nolan A, Frias ES, Grue K, Becker M, Ureta G, Delgado L, Bernales S, Sohal VS, Walter P, Rosi S, Integrated Stress Response Inhibitor Reverses Sex-Dependent Behavioral and Cell-Specific Deficits after Mild Repetitive Head Trauma, J Neurotrauma, 37 (2020) 1370–1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Sidrauski C, Acosta-Alvear D, Khoutorsky A, Vedantham P, Hearn BR, Li H, Gamache K, Gallagher CM, Ang KK, Wilson C, Okreglak V, Ashkenazi A, Hann B, Nader K, Arkin MR, Renslo AR, Sonenberg N, Walter P, Pharmacological brake-release of mRNA translation enhances cognitive memory, Elife, 2 (2013) e00498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Sidrauski C, McGeachy AM, Ingolia NT, Walter P, The small molecule ISRIB reverses the effects of eIF2alpha phosphorylation on translation and stress granule assembly, Elife, 4 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Brown DG, Shorter J, Wobst HJ, Emerging small-molecule therapeutic approaches for amyotrophic lateral sclerosis and frontotemporal dementia, Bioorg Med Chem Lett, 30 (2020) 126942. [DOI] [PubMed] [Google Scholar]
  • [224].Wobst HJ, Mack KL, Brown DG, Brandon NJ, Shorter J, The clinical trial landscape in amyotrophic lateral sclerosis-Past, present, and future, Med Res Rev, 40 (2020) 1352–1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Vigh L, Literati PN, Horvath I, Torok Z, Balogh G, Glatz A, Kovacs E, Boros I, Ferdinandy P, Farkas B, Jaszlits L, Jednakovits A, Koranyi L, Maresca B, Bimoclomol: a nontoxic, hydroxylamine derivative with stress protein-inducing activity and cytoprotective effects, Nat Med, 3 (1997) 1150–1154. [DOI] [PubMed] [Google Scholar]
  • [226].Hargitai J, Lewis H, Boros I, Rácz T, Fiser A, Kurucz I, Benjamin I, Vígh L, Pénzes Z, Csermely P, Latchman DS, Bimoclomol, a heat shock protein co-inducer, acts by the prolonged activation of heat shock factor-1, Biochem Biophys Res Commun, 307 (2003) 689–695. [DOI] [PubMed] [Google Scholar]
  • [227].Kieran D, Kalmar B, Dick JR, Riddoch-Contreras J, Burnstock G, Greensmith L, Treatment with arimoclomol, a coinducer of heat shock proteins, delays disease progression in ALS mice, Nat Med, 10 (2004) 402–405. [DOI] [PubMed] [Google Scholar]
  • [228].Ahmed M, Machado PM, Miller A, Spicer C, Herbelin L, He J, Noel J, Wang Y, McVey AL, Pasnoor M, Gallagher P, Statland J, Lu CH, Kalmar B, Brady S, Sethi H, Samandouras G, Parton M, Holton JL, Weston A, Collinson L, Taylor JP, Schiavo G, Hanna MG, Barohn RJ, Dimachkie MM, Greensmith L, Targeting protein homeostasis in sporadic inclusion body myositis, Sci Transl Med, 8 (2016) 331ra341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Benatar M, Wuu J, Andersen PM, Atassi N, David W, Cudkowicz M, Schoenfeld D, Randomized, double-blind, placebo-controlled trial of arimoclomol in rapidly progressive SOD1 ALS, Neurology, 90 (2018) e565–e574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Mandrioli J, Crippa V, Cereda C, Bonetto V, Zucchi E, Gessani A, Ceroni M, Chio A, D’Amico R, Monsurrò MR, Riva N, Sabatelli M, Silani V, Simone IL, Sorarù G, Provenzani A, D’Agostino VG, Carra S, Poletti A, Proteostasis and ALS: protocol for a phase II, randomised, double-blind, placebo-controlled, multicentre clinical trial for colchicine in ALS (Co-ALS), BMJ Open, 9 (2019) e028486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Crippa V, Carra S, Rusmini P, Sau D, Bolzoni E, Bendotti C, De Biasi S, Poletti A, A role of small heat shock protein B8 (HspB8) in the autophagic removal of misfolded proteins responsible for neurodegenerative diseases, Autophagy, 6 (2010) 958–960. [DOI] [PubMed] [Google Scholar]
  • [232].Crippa V, Sau D, Rusmini P, Boncoraglio A, Onesto E, Bolzoni E, Galbiati M, Fontana E, Marino M, Carra S, Bendotti C, De Biasi S, Poletti A, The small heat shock protein B8 (HspB8) promotes autophagic removal of misfolded proteins involved in amyotrophic lateral sclerosis (ALS), Hum Mol Genet, 19 (2010) 3440–3456. [DOI] [PubMed] [Google Scholar]
  • [233].Ganassi M, Mateju D, Bigi I, Mediani L, Poser I, Lee HO, Seguin SJ, Morelli FF, Vinet J, Leo G, Pansarasa O, Cereda C, Poletti A, Alberti S, Carra S, A Surveillance Function of the HSPB8-BAG3-HSP70 Chaperone Complex Ensures Stress Granule Integrity and Dynamism, Mol Cell, 63 (2016) 796–810. [DOI] [PubMed] [Google Scholar]
  • [234].Anagnostou G, Akbar MT, Paul P, Angelinetta C, Steiner TJ, de Belleroche J, Vesicle associated membrane protein B (VAPB) is decreased in ALS spinal cord, Neurobiol Aging, 31 (2010) 969–985. [DOI] [PubMed] [Google Scholar]
  • [235].Crippa V, Cicardi ME, Ramesh N, Seguin SJ, Ganassi M, Bigi I, Diacci C, Zelotti E, Baratashvili M, Gregory JM, Dobson CM, Cereda C, Pandey UB, Poletti A, Carra S, The chaperone HSPB8 reduces the accumulation of truncated TDP-43 species in cells and protects against TDP-43-mediated toxicity, Hum Mol Genet, 25 (2016) 3908–3924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Soniat M, Chook YM, Nuclear localization signals for four distinct karyopherin-β nuclear import systems, Biochem J, 468 (2015) 353–362. [DOI] [PubMed] [Google Scholar]
  • [237].Nishimura AL, Zupunski V, Troakes C, Kathe C, Fratta P, Howell M, Gallo JM, Hortobágyi T, Shaw CE, Rogelj B, Nuclear import impairment causes cytoplasmic trans-activation response DNA-binding protein accumulation and is associated with frontotemporal lobar degeneration, Brain, 133 (2010) 1763–1771. [DOI] [PubMed] [Google Scholar]
  • [238].Guo L, Fare CM, Shorter J, Therapeutic Dissolution of Aberrant Phases by Nuclear-Import Receptors, Trends Cell Biol, 29 (2019) 308–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Hofweber M, Hutten S, Bourgeois B, Spreitzer E, Niedner-Boblenz A, Schifferer M, Ruepp MD, Simons M, Niessing D, Madl T, Dormann D, Phase Separation of FUS Is Suppressed by Its Nuclear Import Receptor and Arginine Methylation, Cell, 173 (2018) 706–719 e713. [DOI] [PubMed] [Google Scholar]
  • [240].Niaki AG, Sarkar J, Cai X, Rhine K, Vidaurre V, Guy B, Hurst M, Lee JC, Koh HR, Guo L, Fare CM, Shorter J, Myong S, Loss of Dynamic RNA Interaction and Aberrant Phase Separation Induced by Two Distinct Types of ALS/FTD-Linked FUS Mutations, Mol Cell, 77 (2020) 82–94 e84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Rhine K, Makurath MA, Liu J, Skanchy S, Lopez C, Catalan KF, Ma Y, Fare CM, Shorter J, Ha T, Chemla YR, Myong S, ALS/FTLD-Linked Mutations in FUS Glycine Residues Cause Accelerated Gelation and Reduced Interactions with Wild-Type FUS, Mol Cell, 80 (2020) 666–681 e668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Springhower CE, Rosen MK, Chook YM, Karyopherins and condensates, Curr Opin Cell Biol, 64 (2020) 112–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Yoshizawa T, Ali R, Jiou J, Fung HYJ, Burke KA, Kim SJ, Lin Y, Peeples WB, Saltzberg D, Soniat M, Baumhardt JM, Oldenbourg R, Sali A, Fawzi NL, Rosen MK, Chook YM, Nuclear Import Receptor Inhibits Phase Separation of FUS through Binding to Multiple Sites, Cell, 173 (2018) 693–705.e622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Levin AA, Treating Disease at the RNA Level with Oligonucleotides, N Engl J Med, 380 (2019) 57–70. [DOI] [PubMed] [Google Scholar]
  • [245].Ly CV, Miller TM, Emerging antisense oligonucleotide and viral therapies for amyotrophic lateral sclerosis, Curr Opin Neurol, 31 (2018) 648–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Leavitt BR, Tabrizi SJ, Antisense oligonucleotides for neurodegeneration, Science, 367 (2020) 1428–1429. [DOI] [PubMed] [Google Scholar]
  • [247].Apicco DJ, Ash PEA, Maziuk B, LeBlang C, Medalla M, Al Abdullatif A, Ferragud A, Botelho E, Ballance HI, Dhawan U, Boudeau S, Cruz AL, Kashy D, Wong A, Goldberg LR, Yazdani N, Zhang C, Ung CY, Tripodis Y, Kanaan NM, Ikezu T, Cottone P, Leszyk J, Li H, Luebke J, Bryant CD, Wolozin B, Reducing the RNA binding protein TIA1 protects against tau-mediated neurodegeneration in vivo, Nat Neurosci, 21 (2018) 72–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Vanderweyde T, Apicco DJ, Youmans-Kidder K, Ash PEA, Cook C, Lummertz da Rocha E, Jansen-West K, Frame AA, Citro A, Leszyk JD, Ivanov P, Abisambra JF, Steffen M, Li H, Petrucelli L, Wolozin B, Interaction of tau with the RNA-Binding Protein TIA1 Regulates tau Pathophysiology and Toxicity, Cell Rep, 15 (2016) 1455–1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Miller T, Cudkowicz M, Shaw PJ, Andersen PM, Atassi N, Bucelli RC, Genge A, Glass J, Ladha S, Ludolph AL, Maragakis NJ, McDermott CJ, Pestronk A, Ravits J, Salachas F, Trudell R, Van Damme P, Zinman L, Bennett CF, Lane R, Sandrock A, Runz H, Graham D, Houshyar H, McCampbell A, Nestorov I, Chang I, McNeill M, Fanning L, Fradette S, Ferguson TA, Phase 1–2 Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS, N Engl J Med, 383 (2020) 109–119. [DOI] [PubMed] [Google Scholar]
  • [250].Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, Wild EJ, Saft C, Barker RA, Blair NF, Craufurd D, Priller J, Rickards H, Rosser A, Kordasiewicz HB, Czech C, Swayze EE, Norris DA, Baumann T, Gerlach I, Schobel SA, Paz E, Smith AV, Bennett CF, Lane RM, I.-H.S.S.T. Phase 1–2a, Targeting Huntingtin Expression in Patients with Huntington’s Disease, N Engl J Med, 380 (2019) 2307–2316. [DOI] [PubMed] [Google Scholar]
  • [251].Minikel EV, Zhao HT, Le J, O’Moore J, Pitstick R, Graffam S, Carlson GA, Kavanaugh MP, Kriz J, Kim JB, Ma J, Wille H, Aiken J, McKenzie D, Doh-Ura K, Beck M, O’Keefe R, Stathopoulos J, Caron T, Schreiber SL, Carroll JB, Kordasiewicz HB, Cabin DE, Vallabh SM, Prion protein lowering is a disease-modifying therapy across prion disease stages, strains and endpoints, Nucleic Acids Res, 48 (2020) 10615–10631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Raymond GJ, Zhao HT, Race B, Raymond LD, Williams K, Swayze EE, Graffam S, Le J, Caron T, Stathopoulos J, O’Keefe R, Lubke LL, Reidenbach AG, Kraus A, Schreiber SL, Mazur C, Cabin DE, Carroll JB, Minikel EV, Kordasiewicz H, Caughey B, Vallabh SM, Antisense oligonucleotides extend survival of prion-infected mice, JCI Insight, 5 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Alarcon-Aris D, Pavia-Collado R, Miquel-Rio L, Coppola-Segovia V, Ferres-Coy A, Ruiz-Bronchal E, Galofre M, Paz V, Campa L, Revilla R, Montefeltro A, Kordower JH, Vila M, Artigas F, Bortolozzi A, Anti-alpha-synuclein ASO delivered to monoamine neurons prevents alpha-synuclein accumulation in a Parkinson’s disease-like mouse model and in monkeys, EBioMedicine, 59 (2020) 102944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [254].Luna E, Decker SC, Riddle DM, Caputo A, Zhang B, Cole T, Caswell C, Xie SX, Lee VMY, Luk KC, Differential alpha-synuclein expression contributes to selective vulnerability of hippocampal neuron subpopulations to fibril-induced toxicity, Acta Neuropathol, 135 (2018) 855–875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Donnelly CJ, Zhang PW, Pham JT, Haeusler AR, Mistry NA, Vidensky S, Daley EL, Poth EM, Hoover B, Fines DM, Maragakis N, Tienari PJ, Petrucelli L, Traynor BJ, Wang J, Rigo F, Bennett CF, Blackshaw S, Sattler R, Rothstein JD, RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention, Neuron, 80 (2013) 415–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Jiang J, Zhu Q, Gendron TF, Saberi S, McAlonis-Downes M, Seelman A, Stauffer JE, Jafar-Nejad P, Drenner K, Schulte D, Chun S, Sun S, Ling SC, Myers B, Engelhardt J, Katz M, Baughn M, Platoshyn O, Marsala M, Watt A, Heyser CJ, Ard MC, De Muynck L, Daughrity LM, Swing DA, Tessarollo L, Jung CJ, Delpoux A, Utzschneider DT, Hedrick SM, de Jong PJ, Edbauer D, Van Damme P, Petrucelli L, Shaw CE, Bennett CF, Da Cruz S, Ravits J, Rigo F, Cleveland DW, Lagier-Tourenne C, Gain of Toxicity from ALS/FTD-Linked Repeat Expansions in C9ORF72 Is Alleviated by Antisense Oligonucleotides Targeting GGGGCC-Containing RNAs, Neuron, 90 (2016) 535–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Lagier-Tourenne C, Baughn M, Rigo F, Sun S, Liu P, Li HR, Jiang J, Watt AT, Chun S, Katz M, Qiu J, Sun Y, Ling SC, Zhu Q, Polymenidou M, Drenner K, Artates JW, McAlonis-Downes M, Markmiller S, Hutt KR, Pizzo DP, Cady J, Harms MB, Baloh RH, Vandenberg SR, Yeo GW, Fu XD, Bennett CF, Cleveland DW, Ravits J, Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration, Proc Natl Acad Sci U S A, 110 (2013) E4530–4539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Nishimura AL, Shum C, Scotter EL, Abdelgany A, Sardone V, Wright J, Lee YB, Chen HJ, Bilican B, Carrasco M, Maniatis T, Chandran S, Rogelj B, Gallo JM, Shaw CE, Allele-specific knockdown of ALS-associated mutant TDP-43 in neural stem cells derived from induced pluripotent stem cells, PLoS One, 9 (2014) e91269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Arnold C, Custom therapies pose huge financial burdens, Nat Med, (2019). [DOI] [PubMed] [Google Scholar]
  • [260].Maharana S, Wang J, Papadopoulos DK, Richter D, Pozniakovsky A, Poser I, Bickle M, Rizk S, Guillén-Boixet J, Franzmann TM, Jahnel M, Marrone L, Chang Y-T, Sterneckert J, Tomancak P, Hyman AA, Alberti S, RNA buffers the phase separation behavior of prion-like RNA binding proteins, Science (New York, N.Y.), 360 (2018) 918–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Garcia-Jove Navarro M, Kashida S, Chouaib R, Souquere S, Pierron G, Weil D, Gueroui Z, RNA is a critical element for the sizing and the composition of phase-separated RNA-protein condensates, Nature Communications, 10 (2019) 3230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Prasad A, Bharathi V, Sivalingam V, Girdhar A, Patel BK, Molecular Mechanisms of TDP-43 Misfolding and Pathology in Amyotrophic Lateral Sclerosis, Frontiers in molecular neuroscience, 12 (2019) 25–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Ishiguro T, Sato N, Ueyama M, Fujikake N, Sellier C, Kanegami A, Tokuda E, Zamiri B, Gall-Duncan T, Mirceta M, Furukawa Y, Yokota T, Wada K, Taylor JP, Pearson CE, Charlet-Berguerand N, Mizusawa H, Nagai Y, Ishikawa K, Regulatory Role of RNA Chaperone TDP-43 for RNA Misfolding and Repeat-Associated Translation in SCA31, Neuron, 94 (2017) 108–124 e107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Ryan VH, Watters S, Amaya J, Khatiwada B, Venditti V, Naik MT, Fawzi NL, Weak binding to the A2RE RNA rigidifies hnRNPA2 RRMs and reduces liquid-liquid phase separation and aggregation, Nucleic Acids Res, 48 (2020) 10542–10554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Girdhar A, Bharathi V, Tiwari VR, Abhishek S, Deeksha W, Mahawar US, Raju G, Singh SK, Prabusankar G, Rajakumara E, Patel BK, Computational insights into mechanism of AIM4-mediated inhibition of aggregation of TDP-43 protein implicated in ALS and evidence for in vitro inhibition of liquid-liquid phase separation (LLPS) of TDP-43(2C)-A315T by AIM4, Int J Biol Macromol, 147 (2020) 117–130. [DOI] [PubMed] [Google Scholar]
  • [266].Duennwald ML, Shorter J, Countering amyloid polymorphism and drug resistance with minimal drug cocktails, Prion, 4 (2010) 244–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Roberts BE, Duennwald ML, Wang H, Chung C, Lopreiato NP, Sweeny EA, Knight MN, Shorter J, A synergistic small-molecule combination directly eradicates diverse prion strain structures, Nat Chem Biol, 5 (2009) 936–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Shorter J, Emergence and natural selection of drug-resistant prions, Mol Biosyst, 6 (2010) 1115–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Shigenaga MK, Hagen TM, Ames BN, Oxidative damage and mitochondrial decay in aging, Proc Natl Acad Sci U S A, 91 (1994) 10771–10778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Jain S, Wheeler JR, Walters RW, Agrawal A, Barsic A, Parker R, ATPase-Modulated Stress Granules Contain a Diverse Proteome and Substructure, Cell, 164 (2016) 487–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Patel A, Malinovska L, Saha S, Wang J, Alberti S, Krishnan Y, Hyman AA, ATP as a biological hydrotrope, Science, 356 (2017) 753–756. [DOI] [PubMed] [Google Scholar]
  • [272].Li Q, Shortreed MR, Wenger CD, Frey BL, Schaffer LV, Scalf M, Smith LM, Global Post-Translational Modification Discovery, J Proteome Res, 16 (2017) 1383–1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Darling AL, Uversky VN, Intrinsic Disorder and Posttranslational Modifications: The Darker Side of the Biological Dark Matter, Front Genet, 9 (2018) 158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Bah A, Forman-Kay JD, Modulation of Intrinsically Disordered Protein Function by Post-translational Modifications, J Biol Chem, 291 (2016) 6696–6705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Hofweber M, Dormann D, Friend or foe-Post-translational modifications as regulators of phase separation and RNP granule dynamics, J Biol Chem, 294 (2019) 7137–7150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Pak CW, Kosno M, Holehouse AS, Padrick SB, Mittal A, Ali R, Yunus AA, Liu DR, Pappu RV, Rosen MK, Sequence Determinants of Intracellular Phase Separation by Complex Coacervation of a Disordered Protein, Mol Cell, 63 (2016) 72–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [277].Shin Y, Brangwynne CP, Liquid phase condensation in cell physiology and disease, Science, 357 (2017). [DOI] [PubMed] [Google Scholar]
  • [278].Andrusiak MG, Sharifnia P, Lyu X, Wang Z, Dickey AM, Wu Z, Chisholm AD, Jin Y, Inhibition of Axon Regeneration by Liquid-like TIAR-2 Granules, Neuron, 104 (2019) 290–304.e298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [279].Chong FP, Ng KY, Koh RY, Chye SM, Tau Proteins and Tauopathies in Alzheimer’s Disease, Cell Mol Neurobiol, 38 (2018) 965–980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Rhoads SN, Monahan ZT, Yee DS, Shewmaker FP, The Role of Post-Translational Modifications on Prion-Like Aggregation and Liquid-Phase Separation of FUS, Int J Mol Sci, 19 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Murray DT, Kato M, Lin Y, Thurber KR, Hung I, McKnight SL, Tycko R, Structure of FUS Protein Fibrils and Its Relevance to Self-Assembly and Phase Separation of Low-Complexity Domains, Cell, 171 (2017) 615–627.e616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Brady OA, Meng P, Zheng Y, Mao Y, Hu F, Regulation of TDP-43 aggregation by phosphorylation and p62/SQSTM1, J Neurochem, 116 (2011) 248–259. [DOI] [PubMed] [Google Scholar]

RESOURCES