- Review
- Open access
- Published:
Dual modulation of amyloid beta and tau aggregation and dissociation in Alzheimer’s disease: a comprehensive review of the characteristics and therapeutic strategies
Translational Neurodegeneration volume 14, Article number: 15 (2025)
Abstract
Alzheimer’s disease (AD) is not a single-cause disease; rather, it is a complex neurodegenerative disease involving multiple pathological pathways influenced by various risk factors. Aggregation and accumulation of amyloid beta (Aβ) and tau are the most prominent features in the brains of AD patients. Aggregated Aβ and tau exert neurotoxic effects in the central nervous system, contributing to the pathogenesis and progression of AD. They also act synergistically to cause neurodegeneration, resulting in memory loss. In this context, dual inhibition of Aβ and tau aggregation, or dissociation of these two aggregates, is considered promising for AD treatment. Recently, dual inhibitors capable of simultaneously targeting the aggregation and dissociation of both Aβ and tau have been investigated. Specific amino acid domains of Aβ and tau associated with their aggregation/dissociation have been identified. Subsequently, therapeutic agents that prevent aggregation or promote disaggregation by targeting these domains have been identified/developed. In this review, we summarize the major domains and properties involved in Aβ and tau aggregation, as well as the therapeutic effects and mechanisms of agents that simultaneously regulate their aggregation and dissociation. This comprehensive review may contribute to the design and discovery of next-generation dual-targeting drugs for Aβ and tau, potentially leading to the development of more effective therapeutic strategies for AD.
Background
Alzheimer’s disease (AD) is a complex neurodegenerative disease that accounts for the largest proportion of dementia. Over the past century, a variety of AD risk factors, such as sociodemographic, genetic, lifestyle, and environmental factors, as well as physical and mental health conditions, have been extensively studied [1]. Various neuropathological features, including atrophy, cerebrovascular disruption, and proteinopathy, such as amyloid beta (Aβ), tau, transactive response DNA binding protein of 43 kDa, and α-synuclein, have been observed in AD brains [2]. Additionally, the accumulation of Aβ and tau follows several distinct trajectories [3, 4]. However, the most prominent neuropathological characteristics of AD among various types of dementia are the presence of extracellular Aβ plaques and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau proteins in the brain [5]. Aβ peptides are produced through the amyloidogenic process, in which amyloid precursor protein (APP) is sequentially cleaved by β-secretase and γ-secretase while bypassing α-secretase [6]. Tau protein becomes abnormally hyperphosphorylated due to an imbalance in enzymes that add or remove phosphate [7]. Accumulation of Aβ peptides and hyperphosphorylated tau occurs ~ 20 years before AD onset [8, 9], influencing various AD-associated pathologies. Aβ also interacts with tau to exacerbate AD progression through multiple synergistic mechanisms [10,11,12,13,14]. In particular, studies in animal models have demonstrated that the Aβ − tau interactions not only accelerate tau pathology but also exacerbate neuronal damage by impairing neurotransmission, disrupting calcium homeostasis, and destabilizing microtubules [15,16,17,18,19,20,21,22]. Interestingly, Aβ pathology is closely associated with the spread of tau pathology [23], especially in its initial regions within the rhinal cortex. In patients with a higher accumulation of Aβ (specifically, an amyloid burden > 40 centiloid), tau pathology is more likely to extend into the neocortex compared to those with lower Aβ deposition [24]. The spread of tau pathology facilitated by Aβ is a better predictor of cognitive performance of AD patients than Aβ pathology [25]. Several studies have proposed the mechanisms of the interactions between Aβ and tau in the AD brain. First, Aβ not only induces hyperphosphorylation of tau but also promotes the spread of tau aggregates to neuritic plaques. This links the spatial and temporal progression of Aβ and tau pathology [11, 17, 26]. Second, Aβ and tau interact through the long-range neural network that connects the neocortex to the entorhinal cortex, an early site of tau neurofibrillary tangle formation, driving initial tau spreading through long-range neural networks [27]. In the late stages of AD, Aβ strongly interacts with tau in the inferior temporal gyrus to accelerate connectivity-based tau propagation to a wide range of neocortical regions [27]. Finally, interactions between Aβ and tau may involve specific pathways, such as FGFR3 signaling which facilitates tau uptake and aggregation [28], and the norepinephrine signaling pathway, in which Aβ oligomers activate glycogen synthase kinase 3β (GSK3β) via the α2A adrenergic receptor, leading to tau hyperphosphorylation [29]. The cascade of events involving the aggregation and accumulation of Aβ and tau represents one of the crucial trajectories in AD pathogenesis. Therefore, it is necessary to understand the mechanisms of Aβ and tau aggregation and to identify promising therapeutic strategies targeting their aggregates.
Interestingly, Aβ and tau exhibit similar biophysical characteristics and aggregation kinetics; most notably, both form aggregates featuring a common β-sheet structure which exerts neurotoxic effects on the central nervous system [30]. These aggregates are typically formed either through primary nucleation, in which monomers aggregate without the contribution of preformed aggregates, or through secondary nucleation, in which preformed aggregates of the same monomer type catalyze the nucleation from monomers [31, 32]. Among the different forms of Aβ and tau, oligomers and protofibrils are more harmful and damaging to neuronal function than the monomeric form [33, 34]. Moreover, Aβ and tau aggregates synergistically interact to maximize their neurotoxicity [35]. In particular, Aβ aggregates interact with tau to produce toxic effects, while phosphorylated tau promotes Aβ-induced damage to mitochondria in healthy neurons [36]. As both the aggregation of Aβ/tau and their interaction play crucial roles in different aspects of AD pathologies, development of drugs that can simultaneously inhibit Aβ and tau aggregation by targeting common structural elements, such as the β-sheet, is of significant interest.
Notably, several studies have explored the potential of dual inhibitors of Aβ and tau aggregation and dual modulators that can dissociate Aβ and tau aggregates [37,38,39,40,41,42]. In addition, multiple preclinical and clinical trials have investigated the utility of Aβ and tau dual-targeting drugs in the treatment of AD. These include dual Aβ/tau oligomer inhibitors (Takeda Pharmaceuticals and TREVENTIS™), AS-603 (Amyloid Solution), AS-701 (Amyloid Solution), L&J-AD (L & J Bio.), and BEY-2153 (BEYONDBIO; clinical trial No. NCT04476303). BEY-2153 simultaneously targets Aβ and tau by inhibiting the hyperphosphorylation of both tau protein and APP. Interestingly, AS-603 and AS-701, developed by Amyloid Solution, have been reported to target Aβ and tau aggregates by promoting the degradation of both Aβ and tau oligomers and plaques. Preclinical and clinical studies of dual Aβ and tau modulators have shown that a dual regulatory approach to inhibiting Aβ and tau aggregation or dissociating Aβ and tau aggregates may represent an effective strategy for the treatment of AD [37,38,39,40,41,42].
In this review, we describe the aggregation properties of Aβ and tau and explore aggregation-related sites and mechanisms, focusing on potential targets for dual intervention. In addition, we summarize and discuss recent discoveries on dual-targeting modulators of Aβ and tau, including both aggregation inhibitors and aggregate dissociators, which have been shown to attenuate various AD-related pathologies. Finally, we discuss promising therapeutic strategies that involve targeting the modulation of Aβ and tau aggregation/disaggregation, highlighting their potential in the treatment of AD.
Characteristics of Aβ and tau aggregation
Abnormal accumulation of aggregated Aβ and tau is a primary cause of AD. Studies have revealed specific segments in Aβ and tau that are responsible for their aggregation (Fig. 1). Below, we will comprehensively review the mechanisms that lead to the aggregation of Aβ and tau.
Specific regions involved in Aβ and tau aggregation. Within the Aβ peptide sequence, specific hydrophobic segments, including 17LVF19, 32IGL34, and 41IA42, induce self-aggregation. The 17LVFFAED23 segment and the 30AIIGLMV36 segment induce formation of the β-sheet structure of Aβ fibrils. Within the microtubule-binding domain of tau protein, the 275VQIINK280 and 306VQIVYK311 segments promote tau filaments to self-aggregate and form helical structures. The cross-seeding process between Aβ and tau is facilitated by specific interactions. In Aβ, the lysine (K) 16, valine (V) 18, and glutamic acid (E) 22 residues are key players in aggregation. For tau, the 275VQIINK280 and 306VQIVYK311 segments are crucial for aggregation
Mechanisms underlying Aβ aggregation and potential therapeutic strategies for modulating Aβ aggregation
Excessive accumulation of Aβ due to an imbalance between Aβ production and clearance is a critical event in the pathogenesis of AD [43]. Interestingly, studies on familial AD associated with PSEN1 gene mutations have demonstrated a complex pattern of Aβ production: a small number of patients exhibit increased Aβ production, while a large number of patients show decreased Aβ production [44,45,46]. Surprisingly, a comprehensive analysis of 138 PSEN1 mutations causing familial AD revealed that only 10 mutations increase Aβ production, while the others either decrease or have no significant effect on Aβ levels [46]. For example, P264L and P267S mutations reduce the processing efficiency of γ-secretase by interfering with PSEN1 self-cleavage, resulting in reduced production of Aβ [44]. Although the overall production is decreased, the production ratio of longer Aβ species such as Aβ43 and Aβ45 is increased compared to that in healthy controls, whereas the production ratio of shorter Aβ species such as Aβ38 and Aβ40 is decreased. This finding was unaffected by the characteristics and specific site of the PSEN1 mutations [45]. Thus, it is increasingly recognized that Aβ accumulation in AD pathology can be caused by impaired Aβ clearance mechanisms even without Aβ overproduction. Aβ initially exists as monomers in a random coil or an α-helical conformation and progressively adopts β-sheet-rich structures during aggregation into oligomers, protofibrils, fibrils, and amyloid plaques [47]. Although certain monomeric forms of Aβ can transiently adopt β-sheet structures under experimental conditions such as multinuclear NMR (nuclear magnetic resonance) spectroscopy, this is not the predominant conformational state of monomeric Aβ. During the aggregation process of Aβ, specific hydrophobic segments of Aβ, such as 17LVF19, 32IGL34, and 41IA42, play crucial roles in promoting self-assembly. In particular, Aβ42 exhibits higher hydrophobicity and stronger aggregation properties than Aβ40, as it has two additional residues at the C-terminal, isoleucine 41 and alanine 42. This makes Aβ42 the more toxic isoform associated with AD [48]. The isoleucine 41 and alanine 42 residues at the C-terminal promote aggregation and amyloid plaque formation by forming hydrophobic interactions with the C-terminal hydrophobic region of Aβ and between different Aβ peptides. Specifically, the C-terminal hydrophobic region of Aβ42, the 32IGL34 segment, competitively interacts with both the hydrophobic central region, the 17LVF19 segment, and the extreme C-terminal hydrophobic region, the 41IA42 segment. Notably, interaction between the 32IGL34 and the 41IA42 segments results in the exposure of the 17LVF19 segment to solvent molecules. The exposed 17LVF19 segment plays an important role in the intermolecular hydrophobic interactions that drive oligomerization of Aβ42 [49]. Due to the absence of the two C-terminal amino acids 41IA42 and the altered competitive interaction between hydrophobic regions, Aβ40 exhibits a relatively higher probability of interaction between the 32IGL34 and 17LVF19 sections, and, consequently, a lower aggregation tendency than Aβ42 [49]. Indeed, it has been well-established that Aβ42 aggregates faster than Aβ40 due to its higher affinity for self-assembly through hydrophobic interactions [50, 51]. During oligomerization, Aβ preferentially interacts with the 16KLVFFAEDVG25 segment of other Aβ forms [52]. The 17LVFFAED23 segment of the central region and the 30AIIGLMV36 segment of the C-terminal play a crucial role in the formation of the amyloid structure, which is key for the formation of Aβ fibrils [53, 54]. The central and the C-terminal regions form tetramers, each tetramer consisting of two hydrogen-bonded dimers that pack through hydrophobic interactions [55]. Through these aggregation dynamics, Aβ fibrils in the AD brain promote the stacking of Aβ monomers or dimers with similar structures to form amyloid structures, which can further aggregate into fibrils [56,57,58]. Thus, specific Aβ segments, such as 16KLVFFAEDVG25 and 29GAIIGLMVGGVVIA42, significantly contribute to the formation and structural stabilization of Aβ aggregates (Fig. 1).
Considering the aggregation properties of Aβ, diverse compounds can be used to modulate aggregation by targeting specific regions of Aβ. First, Aβ aggregation may be inhibited by an inhibitor binding to the central hydrophobic region of Aβ, the 17LVF19 segment, to form a hydrophobic interaction. Multiple bioactive components extracted from natural products, including catechins, epicatechin, curcumin, and epigallocatechin gallate (EGCG), have been shown to form a hydrophobic interaction with the central hydrophobic region of Aβ [59,60,61,62]. Furthermore, peptide-based drugs with altered structural conformations can interact with the central hydrophobic region of Aβ to reduce aggregation and fibril formation [63,64,65,66,67]. Second, Aβ aggregation may be inhibited through direct interaction with specific regions of the Aβ peptide. For instance, morin and datiscetin target the 10YEVHHQ15 and 19FFA21 sequences of the Aβ42 monomer, subsequently inhibiting the nucleation and elongation phases of amyloid aggregation, protecting neurons from amyloid toxicity [68]. Quercetin recognizes and binds to the 17LVFFA21 sequence of the Aβ oligomer during the early stages of Aβ aggregation [69, 70]. In addition, nicotine and its optical isomers exhibit anti-aggregation effects on Aβ through selective interactions with the histidine residues 6, 13, and 14 in the Aβ peptide [71]. One of the cembranoid compounds derived from the Chinese soft coral Sinularia sp., exerts an anti-aggregation effect on Aβ not only through hydrogen bonding between the OH at C-11 of this compound and the carbonyl of the backbone of Glycine 37 of Aβ42, but also through hydrophobic interactions between the compound and the leucine 34, valine 39, valine 40, isoleucine 41, and alanine 42 residues of Aβ42 monomer [72]. Molecular dynamics simulation further demonstrated that both tanshinone I and tanshinone IIA, compounds isolated from the Chinese herbs Danshen or Salvia Miltiorrhiza Bunge, preferentially bind to a hydrophobic amyloid groove formed by the C-terminal sequences 31IIGLM35 and 35MVGGV39, along with several aromatic residues [73]. Collectively, these results indicate that the specific targeting and binding of key hydrophobic regions of Aβ is an effective approach to modulating Aβ aggregation.
Critical mechanisms of tau aggregation and potential therapeutic strategies for modulating tau aggregation
Tau protein progressively aggregates from monomers to oligomers and paired helical filaments (PHFs), eventually forming NFTs [74]. In the AD brain, abnormally hyperphosphorylated tau dissociates from microtubules, destabilizes the normal structural conformation of microtubules, and has increased propensity to aggregate into pathological forms [75]. This process is mediated by several tau kinases including GSK3β and cyclin-dependent kinase 5, and phosphatases [76]. Hyperphosphorylation of tau occurs primarily in the proline-rich regions and the microtubule-binding domain (MTBD), leading to structural destabilization and exposure of hydrophobic regions that promote aggregation of the tau protein [77]. Aggregation of tau is primarily caused by the hydrophobic interactions between MTBDs, which typically contains three or four repeats [78]. The MTBD has specific aggregation-promoting regions that are predominantly involved in the aggregation of tau. Two hexapeptides, 275VQIINK280 present in the repeat 2 domain (R2), and 306VQIVYK311 present in the repeat 3 domain (R3) of the MTBD, have been well-characterized as aggregation-promoting regions central to tau aggregation [79, 80]. Valine 306 and isoleucine 308 are central for stabilizing the amyloid structure formed by the 275VQIINK280 and the 306VQIVYK311 segments [81]. In particular, the 275VQIINK280 segment in the R2 domain more strongly contributes to the seeding and aggregation than the 306VQIVYK311 segment in the R3 domain [79, 82]. However, neither aggregates of hexapeptide 275VQIINK280 nor aggregates of 306VQIVYK311 show the helical structure observed in PHFs [79]. These results suggest that the two hexapeptides play a critical role in the initial nucleation of tau protein (Fig. 1). In particular, tyrosine 310 of the 306VQIVYK311 peptide has been identified as an essential residue for the formation of these tau filaments [83]. In this region, the tyrosine 310 residue engages in a C–H⋯π interaction with isoleucine 308 to form a steric zipper structure in tau filaments, a typical structure for cross-β-sheet forming peptides [84]. Indeed, the structured core of tau filaments extracted from the brains of AD patients contains the 306VQIVYK311 segment [85]. Tau filaments are composed of two similarly structured protofilaments. The structured core of tau filaments forms a C-shaped architecture comprising eight β-sheets enriched in residues from valine 306 to arginine 406 in the R3 and R4 domains of tau [85, 86]. Importantly, V306–F378, which comprises eight β-sheets, promotes fibrillization and contributes to the stabilization of the fibril structure [87]. Moreover, aggregates of V306–F378 are efficiently internalized through endocytosis, thereby promoting tau propagation [87].
Based on the aggregation properties of tau, many compounds are being developed to modulate tau aggregation to treat AD or tauopathies. First, tau aggregation inhibitors can be covalent; these inhibitors act by forming covalent bonds within residues of the tau protein to prevent aggregation [88]. Methylthioninium chloride, commonly referred to as methylene blue (MB), is a strong inhibitor of tau aggregation [89]. Leuco-methylthioninium bis (LMTX), also known as TRx0237, is a second-generation derivate of MB that binds to abnormal tau and inhibits tau aggregation [90]. MB can modulate tau aggregation by regulating the oxidation of cysteine sulfhydryl groups [91]. In particular, previous studies have shown that the oxidized or reduced forms of MB, known as methylthioninium and LMTX, respectively, bind to the cysteine 291 and cysteine 322 residues of the tau protein, thereby inhibiting tau aggregation [92]. Second, small molecules that interact directly with tau monomers can potentially inhibit the initiation of aggregation by binding to hydrophobic regions. For example, curcumin attenuates tau aggregation through specific molecular interactions. Indeed, structural studies have shown that curcumin binds primarily to the hydrophobic domains of tau, including residues valine 255, valine 292, isoleucine 195, and valine 305. This binding involves not only hydrophobic interactions but also the formation of hydrogen bonds with nearby residues [93]. These hydrophobic interactions and hydrogen bonds both play a critical role in disrupting the steric zipper structure, a key element in tau protein aggregation, thereby inhibiting tau aggregation. Moreover, the rapid change from the exposed state of the hydrophobic region to the normal state reduces the exposure time of the tau hydrophobic residues, thereby reducing the opportunity for interaction with other peptides, subsequently inhibiting aggregation [94]. Finally, tau aggregation may be regulated by blocking the steric zipper structure that forms in cross-β-sheet peptides [88]. In particular, several noncovalent inhibitors of tau aggregation may act by blocking the formation of the steric zipper structures common to cross-β-sheet peptides [88]. Interestingly, some small molecules, including fluorescent dyes, peptide inhibitors D1b, orange G, and curcumin, have been found to disrupt the steric zipper structure across peptides, thus interfering with steric zipper interactions and preventing peptide chains from aggregating into fibrillar structures [88, 95,96,97]. Taken together, these data suggest that specifically targeting the microtubule-binding repeat domains, especially the aggregation-promoting regions, such as the hexapeptides 275VQIINK280 and 306VQIVYK311, through various approaches such as covalent binding, hydrophobic interactions, or disruption of the steric zipper structure, could be an effective strategy to modulate and inhibit tau protein aggregation.
Critical mechanisms underlying the dual modulation of Aβ/tau aggregation and therapeutic agents
Pathologic Aβ and tau act synergistically to deteriorate neurodegeneration in AD [35]. The cross-seeding of misfolded Aβ and tau, in which the misfolded forms of one molecule catalyze the misfolding and aggregation of the other, accelerates AD progression [98]. The interaction between Aβ and tau is mediated by a variety of non-covalent interactions, including: hydrophobic interactions, electrostatic interactions, and hydrogen bonding (Fig. 1). First, hydrophobic interactions cause induction of Aβ peptide and tau protein aggregation. It has been well established that monomers of Aβ can self-assemble into hydrophobic oligomers and fibrils through hydrophobic interactions [99, 100]. In particular, the hydrophobic interaction sites on Aβ not only induce the aggregation of Aβ, but can also interact with tau to enhance the aggregation of both. For example, the hydrophobic residues of Aβ monomer, including tyrosine 10, phenylalanine 20, methionine 35, valine 39, and isoleucine 41, mainly interact with monomeric tau [101]. Furthermore, residues lysine 16, valine 18, and glutamic acid 22 of the Aβ monomer induce tau aggregation and self-seeding when exposed to solvent molecules [102], indicating the involvement of hydrophobic interactions between Aβ and tau. Second, Aβ aggregates interact with tau monomers through electrostatic interactions. The intrinsically disordered tau protein, when aggregated by pro-amyloid factors, adopts an extended conformation that reduces the structural polymorphism of tau and exposes two hexapeptides, 275VQIINK280 of R2 and 306VQIVYK311 of R3 [103]. Aβ aggregates may contain positively charged regions due to the presence of lysine residues or other charged amino acids [104]. The positively charged regions of Aβ aggregates may interact with negatively charged residues, such as glutamine, in the hexapeptide motifs of tau via electrostatic attraction. Moreover, Aβ aggregates or Aβ residues 16–26 bind to the 275VQIINK280 and 306VQIVYK311 of tau to form fibrils with a β-sheet axis [105, 106]. Third, the exposure of hydrogen bond donors and acceptors on Aβ aggregates may affect the simultaneous aggregation of Aβ and tau by forming hydrogen bonds with polar residues present in the hexapeptide motifs of tau, such as glutamine and asparagine [101]. Surprisingly, aggregated Aβ, rather than monomeric Aβ, strongly accelerates the aggregation and propagation of tau both in vitro and in vivo [20, 107, 108]. Taken together, these results suggest that the cross-talk between Aβ and tau plays a critical role in AD-associated pathologies, demonstrating the need for therapeutic strategies to disrupt these deleterious interactions.
As described above, simultaneous aggregation of Aβ and tau is primarily driven by hydrophobic interactions, electrostatic interactions, and hydrogen bonds formed by specific residues between them. Hence, dual inhibitors should interact with the key residues involved in the aggregation of Aβ and tau to prevent aggregate formation by disrupting non-covalent interactions, such as hydrophobic interactions, electrostatic interactions, and hydrogen bonding between Aβ and tau [109, 110]. Some agents can regulate Aβ and tau aggregation simultaneously (Table 1). Phenylindane, a primary component of coffee, inhibits the fibrillization of both Aβ and tau [111]. Curcumin has been extensively studied as a simultaneous regulator of Aβ and tau aggregation. Curcumin binds directly to Aβ and tau monomers, stabilizes oligomeric species, and blocks dehydrogenation potentially involved in Aβ and tau interactions [112,113,114,115]. Additionally, curcumin derivatives simultaneously inhibit the aggregation of Aβ and tau by blocking hydrophobic interaction and development of the amyloid structure [38, 88, 116]. In addition to curcumin, derivatives of tacrine [39], N-benzylpiperidine [37], benzylamine-hydroxyalkylamine [117], and thiophene [118] have also been reported to exert a dual inhibitory activity on Aβ and tau. These dual inhibitors inhibit the exposure of hydrophobic regions present in Aβ and tau monomers or have hydrophobic/electrostatic interactions with the exposed hydrophobic regions, thereby attenuating aggregation. Furthermore, several agents, including Uncaria rhynchophylla [40], genipin and pyrogallol [41], neferine [42], benzylamine-hydroxyalkylamine derivatives [117], necrostatin-1 derivatives [119], curcumin [38, 93, 116, 120], and EGCG [61, 121,122,123,124], not only inhibit the aggregation of Aβ and tau but also promote the disaggregation. In particular, genipin and pyrogallol have sporadic interactions through covalent/non-covalent bonds with the amino acid residues that play a critical role in Aβ and tau fibril formation, thereby regulating aggregation or inducing fibril dissociation [41]. Moreover, curcumin, EGCG, and Nec1-derivates inhibit Aβ aggregation and promote the dissociation of Aβ protofibrils by breaking the hydrogen bond [119]. Conversely, EGCG regulates the aggregation of Aβ by forming new π-π interactions with histidine 6 and glutamic acid 11 and forming a hydrogen bond with histidine 14/tyrosine 10 and a hydrogen bond with glutamic acid 11 [124]. EGCG also regulates Aβ aggregation by disrupting the lysine 28–alanine 42 salt bridge through a hydrogen bond with Aβ42 as well as the cation-π interaction between its gallic acid ester group and lysine 28 [124]. Furthermore, neferine, curcumin, and EGCG suppress tau aggregation and promote tau dissociation [122]. In particular, EGCG stacks in the polar gap between paired helical filaments. Specifically, EGCG promotes the early disassembly of tau fibrils by competing with the hydrogen bonds, specifically at the serine 341 residue, that maintain tau molecules together in tau fibrils [123]. In addition, neferine, curcumin, and EGCG induce a conformational change from a β-sheet to an unfolded monomeric form, which inhibits aggregation and promotes degradation of Aβ and tau [42, 122]. In this regard, various kinetic studies have demonstrated that the structural disorder of Aβ and tau is crucial for aggregation [125,126,127]. Unfortunately, among the various interactions that contribute to the anti-aggregation and the degradation effects of dual modulators (such as genipin, pyrogllol, neferine, curcumin, and EGCG) on Aβ and tau, the most important interactions remain unclear. However, the different interactions of the molecules with both Aβ and tau may act in a complementary and synergistic manner. The complementary action may enhance the overall efficacy of dual modulators by targeting multiple interactions involved in Aβ/tau aggregation and disaggregation, providing a more robust mechanism to inhibit or dissociate pathological aggregate formation. Taken together, several approaches could be employed to inhibit Aβ and tau aggregation while promoting their dissociation: (1) inhibiting the aggregation of Aβ and tau by preventing the exposure of hydrophobic regions on Aβ and tau monomers, or through interactions with these regions to weaken areas prone to aggregation; and (2) promoting the disassembly of Aβ and tau by inducing sporadic interactions through covalent or non-covalent bonds with amino acid residues critical for fibrillation (Fig. 2).
Possible mechanisms of action of dual modulators of Aβ and tau aggregation and their aggregate dissociation. These dual inhibitors of Aβ and tau act (1) to inhibit aggregation by preventing exposure of the hydrophobic regions of Aβ and tau monomers, or by inducing interactions with hydrophobic regions to weaken vulnerable regions of Aβ and tau, and (2) to modulate aggregation by inducing sporadic interactions through covalent or non-covalent bonds with amino acid residues important for Aβ and tau fibrillation. The dual modulators for Aβ and tau aggregate dissociation would induce sporadic interactions through covalent or non-covalent bonds with amino acid residues that are important for the fibrillation of Aβ and tau, leading to dissociation of the fibrils
In addition to the dual modulators such as pyrogallol, genipin, neferine, and EGCG, which have known mechanisms for regulating Aβ and tau aggregation, multiple other agents have been proposed to target Aβ and tau aggregation. Carbon dots, a type of carbon-based nanomaterials, have gained significant attention for their therapeutic effects on AD pathology. Interestingly, carbon dots can directly interact with Aβ peptides to inhibit Aβ misfolding and aggregation [128] and prevent tau aggregation [129]. Furthermore, carbon dots derived from congo red have the potential to simultaneously inhibit Aβ and tau aggregation [130]. Molecular tweezers are small molecules designed to bind to specific sites on target proteins, thereby preventing abnormal protein aggregation, such as amyloid-associated deposits [131]. The molecular tweezers can reduce Aβ plaques, NFTs, and microgliosis through modulating Aβ and tau aggregation in 3×Tg mice [132]. Similarly, there has been extensive research on structure-based peptide inhibitors that bind to specific sequences in Aβ and tau [82, 102, 133, 134]. These structure-based inhibitors can suppress the aggregation of Aβ and tau and inhibit their toxicity. Interestingly, congo red is proposed as one of the candidate dual modulators of Aβ and tau aggregation [130]. Congo red is a water-soluble diazo dye commonly used to detect Aβ in AD. It can bind to amyloid aggregates, particularly the β-sheet structure of amyloid fibrils. A study demonstrated that congo red can inhibit Aβ and tau aggregation in vitro, implying a significant potential for the treatment of amyloid-related diseases [130]. Despite numerous studies, the precise molecular mechanisms underlying the effects of carbon dots, molecular tweezers, and congo red on Aβ and tau aggregation have yet to be fully elucidated.
Advantages and disadvantages of the therapeutic agents
The simultaneous targeting of Aβ and tau offers a multi-faceted therapeutic strategy for the treatment of AD. First, dual targeting of Aβ and tau can prevent the synergistic interaction between them. Studies have shown that Aβ accelerates the aggregation of tau [21, 137] and that tau, in turn, can affect the properties of Aβ plaques [22]. These findings highlight the complex relationship between Aβ and tau, with each molecule exacerbating the aggregation and pathological effects in AD [138]. Moreover, the synergistic effects between Aβ and tau contribute to the exacerbation of AD-related pathologies, including cognitive decline and neurodegeneration [139, 140]. Dual inhibitors targeting both Aβ and tau may not only prevent the formation of Aβ and tau tangles individually but also disrupt the cross-linking, a key mechanism driving the formation of pathological aggregates. Therefore, dual inhibitors targeting both Aβ and tau represent a promising therapeutic approach to mitigating AD pathology by addressing both individual and synergistic mechanisms. Second, multi-target therapeutic approaches can significantly reduce the probability of resistance. Single-target strategies are often limited since proteins can evolve to become resistant to drugs that block specific mechanisms, reducing single-target drug efficacy over time [141]. The simultaneous modulation of Aβ and tau aggregation better reflects the complexity of the biological system and may be more effective in slowing AD progression. Third, dual targeting of Aβ and tau may offer the potential to design therapies specific to a personalized biological profile, in line with the concept of precision medicine. The aggregation patterns of Aβ and tau differ among individuals due to genetic, age-related, and environmental factors [142]. Susceptibility to aggregation varies among patients with AD. For example, one subgroup may have a higher propensity for Aβ aggregation and another subgroup for tau aggregation. Therefore, simultaneous modulation of Aβ and tau represents an innovative approach for AD treatment with the potential to reduce treatment resistance and meanwhile enable personalized treatment.
Despite the promising benefits, there are potential disadvantages to the use of therapeutic agents for inhibiting Aβ/tau aggregation and dissociating their aggregates. The first concern is the specificity and selectivity of Aβ/tau aggregation modulators. The specificity of modulators for Aβ and tau aggregation varies depending on the target regions involved in Aβ and tau aggregation. Aβ modulators target critical hydrophobic regions at which Aβ self-assembles into pathogenic aggregates. Modulators of Aβ also disrupt the sheet-forming segments, interfering with the molecular mechanisms of Aβ aggregation. Similarly, modulators of tau aggregation specifically target repeat domains within the MTBD, characterized by prominent β-sheet-forming and hydrophobic regions that are critical for tau aggregation. However, dual modulators for Aβ and tau aggregation may target and affect common structural elements such as hydrophobic cores and β-sheet regions, which may be present in other proteins, including α-synuclein and prions, and may cause detrimental effects on protein function [143, 144]. Disruption of physiological proteins through non-specific interactions could disrupt cytoskeletal organization, impair synaptic integrity, or dysregulate energy homeostasis, ultimately exacerbating neuronal dysfunction. It can also disrupt protein homeostasis and protein–protein interactions [145, 146]. Dual modulators of Aβ and tau aggregation may provide a safer alternative with fewer side effects compared to anti-amyloid monoclonal antibodies (MABs). While anti-amyloid MABs, such as aducanumab, lecanemab, and donanemab, have demonstrated efficacy in targeting Aβ, they are also associated with a higher risk of amyloid-related imaging abnormalities, brain atrophy, and inflammatory responses, which can result in parenchymal edema and microbleeds [147, 148]. On the other hand, Aβ and tau aggregation modulators have potentially fewer risks and side effects by preventing broad activation of the immune system, compared with anti-amyloid MABs.
The second concern is that dual modulators of Aβ and tau may show heterogeneous therapeutic efficacy in different pathological subtypes of AD. One key challenge is identifying which patients will benefit most from dual inhibitors of Aβ and tau, especially considering that not all AD subtypes exhibit similar levels of Aβ and tau accumulation in the brain. APOE ε4 carriers typically exhibit higher Aβ burden in the neocortex and hippocampus and increased tau deposition in the medial and lateral temporal cortices, compared to non-carriers [149, 150]. In addition, a study of molecular heterogeneity in AD patients defined at least three different molecular subtypes of AD [151]. As a result of the different subtypes of AD, it may be difficult to predict the clinical efficacy of new inhibitors. However, importantly, current guidelines for AD clinical trials prioritize selecting patients based on biomarkers, such as Aβ and tau [152]. The primary candidates for treatment are those who present with such biomarker profiles, regardless of AD subtype. In addition, both familial and sporadic AD patients undergo accumulation of Aβ, followed by tau, ~ 20 years prior to the onset of AD symptoms [8, 9]. Although it is hard to ensure equal therapeutic efficacy across all subtypes of AD, such a dual-inhibitor approach represents the most promising therapeutic strategy to benefit the widest range of AD subtypes, particularly considering the biomarker-driven patient selection criteria and the fundamental role of both Aβ and tau in the pathogenesis of AD.
The final concern is whether the Aβ/tau aggregation modulators can cross the blood–brain barrier (BBB) and exert therapeutic efficacy for AD pathology. BBB penetration is of considerable concern for treating neurodegenerative diseases such as AD. Unfortunately, numerous tau aggregation modulators identified through in vitro studies have properties, such as permanent cations, that prevent them from crossing the BBB effectively, which limits their efficacy [153]. There is a need for new approaches to improve drug delivery across the BBB which facilitates the transport of Aβ and tau modulators to the brain. First, transferrin-conjugated nanoparticles may have improved delivery of Aβ and tau modulators through receptor-mediated transcytosis [154,155,156]. Second, nanobodies can cross the BBB through a variety of mechanisms [157, 158] and can therefore be used to deliver conjugated Aβ/tau modulators into the AD brain. Finally, extracellular vesicles and nanomaterials have gained attention as a potential way to facilitate targeted drug delivery across the BBB [159, 160]. These advanced drug delivery strategies can be used to overcome the challenges of BBB permeability, enabling more effective delivery of Aβ/tau modulators and enhancing their therapeutic potential in the treatment of AD.
Conclusion
Anti-amyloid MABs, including aducanumab (Aduhelm®), lecanemab (Leqembi®) and donanemab (Kisunla®), represent the first generation of disease-modifying therapies for AD and have shown significant clinical benefits in reducing Aβ plaques in the AD brain [161,162,163]. Targeting Aβ aggregation has proven a clinically relevant approach, advancing AD treatment strategies [164, 165]. Research progress in targeting Aβ aggregation has highlighted the need to address the aggregation of other AD-related pathological molecules, driving the development of therapies aimed at both Aβ and tau aggregation. Here, we review the therapeutic agents that can simultaneously regulate the aggregation of both Aβ and tau in AD, as well as their mechanisms of action. Dual modulators of Aβ and tau aggregation can prevent aggregation and promote disaggregation of Aβ and tau in experimental models of AD. Hydrophobic regions, histidine residues, C-terminal regions, and β-sheet grooves of Aβ are important targets of the modulators. Moreover, the microtubule-binding repeat domains of tau, such as the hexapeptides 275VQIINK280 and 306VQIVYK311, are important targets for modulating tau aggregation. In summary, to target Aβ and tau simultaneously, it is important to: (1) prevent the exposure of hydrophobic regions present in Aβ and tau monomers, or induce hydrophobic/electrostatic interactions with the exposed hydrophobic regions, thereby weakening the regions/characteristics involved in aggregation and regulating aggregation; and (2) to induce sporadic interactions through covalent/non-covalent bonds with the amino acid residues involved in the formation of Aβ and tau fibrils, thereby regulating aggregation or inducing fibril dissociation to control aggregation. Nonetheless, although numerous dual inhibitors targeting both Aβ and tau aggregation exist, further research is required to fully understand their binding sites and mechanisms of action. Although inhibition of Aβ peptide and tau protein aggregation has been extensively investigated, therapeutic agents that dissociate their aggregates have been relatively less explored. Nevertheless, it is well known that disruption of the amyloid structure of preformed Aβ and tau aggregates requires a compound with the ability to induce stronger interactions than those that form the β-sheet conformation [166, 167]. However, the discovery of molecules that can mediate sufficiently strong interactions remains a significant challenge.
In conclusion, we review the different sites and mechanisms of action of drugs that regulate Aβ and tau aggregation, suggesting that the development of compounds that dual-target Aβ and tau aggregation based on these mechanisms may be a promising therapeutic strategy for AD.
Availability of data and materials
Not applicable.
Abbreviations
- AD:
-
Alzheimer’s disease
- Aβ:
-
Amyloid beta
- APP:
-
Amyloid precursor protein
- MAB:
-
Anti-amyloid monoclonal antibody
- BBB:
-
Blood-brain barrier
- EGCG:
-
Epigallocatechin gallate
- GSK3β:
-
Glycogen synthase kinase 3β
- LMTX:
-
Leuco-methylthioninium bis
- MB:
-
Methylene blue
- MTBD:
-
Microtubule-binding domain
- NFTs:
-
Neurofibrillary tangles
- PHFs:
-
Paired helical filaments
- R2:
-
Repeat 2 domain
- R3:
-
Repeat 3 domain
References
Livingston G, Huntley J, Liu KY, Costafreda SG, Selbaek G, Alladi S, et al. Dementia prevention, intervention, and care: 2024 report of the Lancet Standing Commission. Lancet. 2024;404(10452):572–628.
DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener. 2019;14(1):32.
Kakuda N, Miyasaka T, Iwasaki N, Nirasawa T, Wada-Kakuda S, Takahashi-Fujigasaki J, et al. Distinct deposition of amyloid-beta species in brains with Alzheimer’s disease pathology visualized with MALDI imaging mass spectrometry. Acta Neuropathol Commun. 2017;5(1):73.
Vogel JW, Young AL, Oxtoby NP, Smith R, Ossenkoppele R, Strandberg OT, et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat Med. 2021;27(5):871–81.
Knopman DS, Amieva H, Petersen RC, Chetelat G, Holtzman DM, Hyman BT, et al. Alzheimer disease. Nat Rev Dis Primers. 2021;7(1):33.
O’Brien RJ, Wong PC. Amyloid precursor protein processing and Alzheimer’s disease. Annu Rev Neurosci. 2011;34:185–204.
Wang JZ, Xia YY, Grundke-Iqbal I, Iqbal K. Abnormal hyperphosphorylation of tau: sites, regulation, and molecular mechanism of neurofibrillary degeneration. J Alzheimers Dis. 2013;33(Suppl 1):S123–39.
Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med. 2012;367(9):795–804.
Jia J, Ning Y, Chen M, Wang S, Yang H, Li F, et al. Biomarker changes during 20 years preceding Alzheimer’s disease. N Engl J Med. 2024;390(8):712–22.
Ittner LM, Götz J. Amyloid-β and tau–a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci. 2011;12(2):65–72.
Zhang H, Wei W, Zhao M, Ma L, Jiang X, Pei H, et al. Interaction between Aβ and Tau in the pathogenesis of Alzheimer’s disease. Int J Biol Sci. 2021;17(9):2181–92.
Rajmohan R, Reddy PH. Amyloid-beta and phosphorylated tau accumulations cause abnormalities at synapses of Alzheimer’s disease neurons. J Alzheimers Dis. 2017;57(4):975–99.
Bloom GS. Amyloid-β and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 2014;71(4):505–8.
Busche MA, Hyman BT. Synergy between amyloid-β and tau in Alzheimer’s disease. Nat Neurosci. 2020;23(10):1183–93.
Clinton L, Blurton-Jones M, Myczek K, Trojanowski J, LaFerla F. Synergistic interactions between Aβ, tau, and α-synuclein: acceleration of neuropathology and cognitive decline. J Neurosci. 2010;30:7281–9.
Manczak M, Reddy PH. Abnormal interaction of oligomeric amyloid-β with phosphorylated tau: implications to synaptic dysfunction and neuronal damage. J Alzheimers Dis. 2013;36(2):285–95.
Hurtado DE, Molina-Porcel L, Iba M, Aboagye AK, Paul SM, Trojanowski JQ, et al. A{beta} accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am J Pathol. 2010;177(4):1977–88.
Pascoal TA, Mathotaarachchi S, Mohades S, Benedet AL, Chung CO, Shin M, et al. Amyloid-β and hyperphosphorylated tau synergy drives metabolic decline in preclinical Alzheimer’s disease. Mol Psychiatry. 2017;22(2):306–11.
Gulisano W, Maugeri D, Baltrons MA, Fà M, Amato A, Palmeri A, et al. Role of amyloid-β and tau proteins in Alzheimer’s disease: confuting the amyloid cascade. J Alzheimers Dis. 2019;68(1):415.
Bolmont T, Clavaguera F, Meyer-Luehmann M, Herzig MC, Radde R, Staufenbiel M, et al. Induction of tau pathology by intracerebral infusion of amyloid-beta-containing brain extract and by amyloid-beta deposition in APP x Tau transgenic mice. Am J Pathol. 2007;171(6):2012–20.
Pooler AM, Polydoro M, Maury EA, Nicholls SB, Reddy SM, Wegmann S, et al. Amyloid accelerates tau propagation and toxicity in a model of early Alzheimer’s disease. Acta Neuropathol Commun. 2015;3:14.
Jackson RJ, Rudinskiy N, Herrmann AG, Croft S, Kim JM, Petrova V, et al. Human tau increases amyloid beta plaque size but not amyloid beta-mediated synapse loss in a novel mouse model of Alzheimer’s disease. Eur J Neurosci. 2016;44(12):3056–66.
Karran E, De Strooper B. The amyloid hypothesis in Alzheimer disease: new insights from new therapeutics. Nat Rev Drug Discov. 2022;21(4):306–18.
Sanchez JS, Becker JA, Jacobs HIL, Hanseeuw BJ, Jiang S, Schultz AP, et al. The cortical origin and initial spread of medial temporal tauopathy in Alzheimer’s disease assessed with positron emission tomography. Sci Transl Med. 2021;13(577):eabc0655.
Brier MR, Gordon B, Friedrichsen K, McCarthy J, Stern A, Christensen J, et al. Tau and Abeta imaging, CSF measures, and cognition in Alzheimer’s disease. Sci Transl Med. 2016;8(338):338ra66.
Pichet Binette A, Franzmeier N, Spotorno N, Ewers M, Brendel M, Biel D, et al. Amyloid-associated increases in soluble tau relate to tau aggregation rates and cognitive decline in early Alzheimer’s disease. Nat Commun. 2022;13(1):6635.
Lee WJ, Brown JA, Kim HR, La Joie R, Cho H, Lyoo CH, et al. Regional Aβ-tau interactions promote onset and acceleration of Alzheimer’s disease tau spreading. Neuron. 2022;110(12):1932-43.e5.
Kim DK, Suh K, Park J, Lee S-E, Han J, Chang S, et al. FGFR3 drives Aβ-induced tau uptake. Exp Mol Med. 2024;56(7):1631–42.
Zhang F, Gannon M, Chen Y, Yan S, Zhang S, Feng W, et al. β-amyloid redirects norepinephrine signaling to activate the pathogenic GSK3β/tau cascade. Sci Transl Med. 2020;12(526):eaay6931.
Malafaia D, Albuquerque HMT, Silva AMS. Amyloid-beta and tau aggregation dual-inhibitors: a synthetic and structure-activity relationship focused review. Eur J Med Chem. 2021;214: 113209.
Rodriguez Camargo DC, Sileikis E, Chia S, Axell E, Bernfur K, Cataldi RL, et al. Proliferation of Tau 304–380 fragment aggregates through autocatalytic secondary nucleation. ACS Chem Neurosci. 2021;12(23):4406–15.
Thacker D, Barghouth M, Bless M, Zhang E, Linse S. Direct observation of secondary nucleation along the fibril surface of the amyloid beta 42 peptide. Proc Natl Acad Sci U S A. 2023;120(25): e2220664120.
Amin L, Harris DA. Abeta receptors specifically recognize molecular features displayed by fibril ends and neurotoxic oligomers. Nat Commun. 2021;12(1):3451.
Gerson JE, Mudher A, Kayed R. Potential mechanisms and implications for the formation of tau oligomeric strains. Crit Rev Biochem Mol Biol. 2016 Nov/Dec;51(6):482–96.
Ittner LM, Gotz J. Amyloid-beta and tau–a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci. 2011;12(2):65–72.
Do TD, Economou NJ, Chamas A, Buratto SK, Shea JE, Bowers MT. Interactions between amyloid-β and tau fragments promote aberrant aggregates: implications for amyloid toxicity. J Phys Chem B. 2014;118(38):11220–30.
Szalaj N, Godyn J, Jonczyk J, Pasieka A, Panek D, Wichur T, et al. Multidirectional in vitro and in cellulo studies as a tool for identification of multi-target-directed ligands aiming at symptoms and causes of Alzheimer’s disease. J Enzyme Inhib Med Chem. 2020;35(1):1944–52.
Okuda M, Fujita Y, Hijikuro I, Wada M, Uemura T, Kobayashi Y, et al. PE859, a novel curcumin derivative, inhibits amyloid-beta and tau aggregation, and ameliorates cognitive dysfunction in senescence-accelerated mouse prone 8. J Alzheimers Dis. 2017;59(1):313–28.
Lv P, Xia CL, Wang N, Liu ZQ, Huang ZS, Huang SL. Synthesis and evaluation of 1,2,3,4-tetrahydro-1-acridone analogues as potential dual inhibitors for amyloid-beta and tau aggregation. Bioorg Med Chem. 2018;26(16):4693–705.
Kim S, Nam Y, Shin SJ, Prajapati R, Shin SM, Kim MJ, et al. Dual modulators of aggregation and dissociation of amyloid beta and tau: In vitro, in vivo, and in silico studies of Uncaria rhynchophylla and its bioactive components. Biomed Pharmacother. 2022;156: 113865.
Kim S, Hyun DG, Nam Y, Shin SJ, Im D, Kim HS, et al. Genipin and pyrogallol: two natural small molecules targeting the modulation of disordered proteins in Alzheimer’s disease. Biomed Pharmacother. 2023;168: 115770.
Nam Y, Prajapati R, Kim S, Shin SJ, Cheong DY, Park YH, et al. Dual regulatory effects of neferine on amyloid-beta and tau aggregation studied by in silico, in vitro, and lab-on-a-chip technology. Biomed Pharmacother. 2024;172: 116226.
Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E, et al. Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol. 2015;11(8):457–70.
Ben-Gedalya T, Moll L, Bejerano-Sagie M, Frere S, Cabral WA, Friedmann-Morvinski D, et al. Alzheimer’s disease-causing proline substitutions lead to presenilin 1 aggregation and malfunction. Embo j. 2015;34(22):2820–39.
Szaruga M, Veugelen S, Benurwar M, Lismont S, Sepulveda-Falla D, Lleo A, et al. Qualitative changes in human gamma-secretase underlie familial Alzheimer’s disease. J Exp Med. 2015;212(12):2003–13.
Sun L, Zhou R, Yang G, Shi Y. Analysis of 138 pathogenic mutations in presenilin-1 on the in vitro production of Aβ42 and Aβ40 peptides by γ-secretase. Proc Natl Acad Sci U S A. 2017;114(4):E476–85.
Drolle E, Hane F, Lee B, Leonenko Z. Atomic force microscopy to study molecular mechanisms of amyloid fibril formation and toxicity in Alzheimer’s disease. Drug Metab Rev. 2014;46(2):207–23.
Im D, Choi TS. Distinctive contribution of two additional residues in protein aggregation of Aβ42 and Aβ40 isoforms. BMB Rep. 2024;57(6):263–72.
Im D, Kim S, Yoon G, Hyun DG, Eom YG, Lee YE, et al. Decoding the roles of amyloid-beta (1–42)’s key oligomerization domains toward designing epitope-specific aggregation inhibitors. JACS Au. 2023;3(4):1065–75.
Jarrett JT, Berger EP, Lansbury PT Jr. The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer’s disease. Biochemistry. 1993;32(18):4693–7.
Hampel H, Shen Y, Walsh DM, Aisen P, Shaw LM, Zetterberg H, et al. Biological markers of amyloid beta-related mechanisms in Alzheimer’s disease. Exp Neurol. 2010;223(2):334–46.
Konstantoulea K, Guerreiro P, Ramakers M, Louros N, Aubrey LD, Houben B, et al. Heterotypic amyloid beta interactions facilitate amyloid assembly and modify amyloid structure. EMBO J. 2022;41(2): e108591.
Itoh SG, Yagi-Utsumi M, Kato K, Okumura H. Key residue for aggregation of amyloid-β peptides. ACS Chem Neurosci. 2022;13(22):3139–51.
Liu R, McAllister C, Lyubchenko Y, Sierks M. Residues 17–20 and 30–35 of beta-amyloid play critical roles in aggregation. J Neurosci Res. 2004;75:162–71.
Truex NL, Wang Y, Nowick JS. Assembly of peptides derived from beta-sheet regions of beta-amyloid. J Am Chem Soc. 2016;138(42):13882–90.
Yang Y, Arseni D, Zhang W, Huang M, Lovestam S, Schweighauser M, et al. Cryo-EM structures of amyloid-beta 42 filaments from human brains. Science. 2022;375(6577):167–72.
Kollmer M, Close W, Funk L, Rasmussen J, Bsoul A, Schierhorn A, et al. Cryo-EM structure and polymorphism of Abeta amyloid fibrils purified from Alzheimer’s brain tissue. Nat Commun. 2019;10(1):4760.
Gremer L, Scholzel D, Schenk C, Reinartz E, Labahn J, Ravelli RBG, et al. Fibril structure of amyloid-beta(1–42) by cryo-electron microscopy. Science. 2017;358(6359):116–9.
Heo HJ, Lee CY. Epicatechin and catechin in cocoa inhibit amyloid beta protein induced apoptosis. J Agric Food Chem. 2005;53(5):1445–8.
Rajasekhar K, Madhu C, Govindaraju T. Natural tripeptide-based inhibitor of multifaceted amyloid beta toxicity. ACS Chem Neurosci. 2016;7(9):1300–10.
Tavanti F, Pedone A, Menziani MC. Insights into the effect of curcumin and (-)-epigallocatechin-3-gallate on the aggregation of Abeta(1–40) monomers by means of molecular dynamics. Int J Mol Sci. 2020;21(15):5462.
Firouzi R, Sowlati-Hashjin S, Chavez-Garcia C, Ashouri M, Karimi-Jafari MH, Karttunen M. Identification of catechins’ binding sites in monomeric Abeta(42) through ensemble docking and MD simulations. Int J Mol Sci. 2023;24(9):8161.
Matsunaga Y, Fujii A, Awasthi A, Yokotani J, Takakura T, Yamada T. Eight-residue Abeta peptides inhibit the aggregation and enzymatic activity of Abeta42. Regul Pept. 2004;120(1–3):227–36.
Tonelli M, Catto M, Tasso B, Novelli F, Canu C, Iusco G, et al. Multitarget therapeutic leads for Alzheimer’s disease: quinolizidinyl derivatives of bi- and tricyclic systems as dual inhibitors of cholinesterases and beta-amyloid (Abeta) aggregation. ChemMedChem. 2015;10(6):1040–53.
Novick PA, Lopes DH, Branson KM, Esteras-Chopo A, Graef IA, Bitan G, et al. Design of beta-amyloid aggregation inhibitors from a predicted structural motif. J Med Chem. 2012;55(7):3002–10.
Yee AW, Aldeghi M, Blakeley MP, Ostermann A, Mas PJ, Moulin M, et al. A molecular mechanism for transthyretin amyloidogenesis. Nat Commun. 2019;10(1):925.
Amijee H, Bate C, Williams A, Virdee J, Jeggo R, Spanswick D, et al. The N-methylated peptide SEN304 powerfully inhibits Abeta(1–42) toxicity by perturbing oligomer formation. Biochemistry. 2012;51(42):8338–52.
Hanaki M, Murakami K, Akagi K, Irie K. Structural insights into mechanisms for inhibiting amyloid beta42 aggregation by non-catechol-type flavonoids. Bioorg Med Chem. 2016;24(2):304–13.
Fang M, Wang X, Su K, Jia X, Guan P, Hu X. Inhibition effect and molecular mechanisms of quercetin on the Abeta42 dimer: a molecular dynamics simulation study. ACS Omega. 2023;8(20):18009–18.
Alghamdi A, Birch DJS, Vyshemirsky V, Rolinski OJ. Impact of the flavonoid quercetin on beta-amyloid aggregation revealed by intrinsic fluorescence. J Phys Chem B. 2022;126(38):7229–37.
Moore SA, Huckerby TN, Gibson GL, Fullwood NJ, Turnbull S, Tabner BJ, et al. Both the D-(+) and L-(-) enantiomers of nicotine inhibit Abeta aggregation and cytotoxicity. Biochemistry. 2004;43(3):819–26.
Jiang CS, Ru T, Yao LG, Miao ZH, Guo YW. Four new cembranoids from the Chinese soft coral Sinularia sp. and their anti-Abeta aggregation activities. Fitoterapia. 2019;136:104176.
Wang Q, Yu X, Patal K, Hu R, Chuang S, Zhang G, et al. Tanshinones inhibit amyloid aggregation by amyloid-beta peptide, disaggregate amyloid fibrils, and protect cultured cells. ACS Chem Neurosci. 2013;4(6):1004–15.
Mamun AA, Uddin MS, Mathew B, Ashraf GM. Toxic tau: structural origins of tau aggregation in Alzheimer’s disease. Neural Regen Res. 2020;15(8):1417–20.
Iqbal K, Alonso Adel C, Chen S, Chohan MO, El-Akkad E, Gong CX, et al. Tau pathology in Alzheimer disease and other tauopathies. Biochim Biophys Acta. 2005;1739(2–3):198–210.
Stoothoff WH, Johnson GVW. Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta. 2005;1739(2–3):280–97.
Rawat P, Sehar U, Bisht J, Selman A, Culberson J, Reddy PH. Phosphorylated tau in Alzheimer’s disease and other tauopathies. Int J Mol Sci. 2022;23(21):12841.
Kolarova M, Garcia-Sierra F, Bartos A, Ricny J, Ripova D. Structure and pathology of tau protein in Alzheimer disease. Int J Alzheimers Dis. 2012;2012: 731526.
von Bergen M, Barghorn S, Li L, Marx A, Biernat J, Mandelkow EM, et al. Mutations of tau protein in frontotemporal dementia promote aggregation of paired helical filaments by enhancing local beta-structure. J Biol Chem. 2001;276(51):48165–74.
von Bergen M, Friedhoff P, Biernat J, Heberle J, Mandelkow EM, Mandelkow E. Assembly of tau protein into Alzheimer paired helical filaments depends on a local sequence motif ((306)VQIVYK(311)) forming beta structure. Proc Natl Acad Sci U S A. 2000;97(10):5129–34.
Mullapudi V, Vaquer-Alicea J, Bommareddy V, Vega AR, Ryder BD, White CL 3rd, et al. Network of hotspot interactions cluster tau amyloid folds. Nat Commun. 2023;14(1):895.
Seidler PM, Boyer DR, Rodriguez JA, Sawaya MR, Cascio D, Murray K, et al. Structure-based inhibitors of tau aggregation. Nat Chem. 2018;10(2):170–6.
Nishiura C, Takeuchi K, Minoura K, Sumida M, Taniguchi T, Tomoo K, et al. Importance of Tyr310 residue in the third repeat of microtubule binding domain for filament formation of tau protein. J Biochem. 2010;147(3):405–14.
Naruto K, Minoura K, Okuda R, Taniguchi T, In Y, Ishida T, et al. Interplay between I308 and Y310 residues in the third repeat of microtubule-binding domain is essential for tau filament formation. FEBS Lett. 2010;584(19):4233–6.
Fitzpatrick AWP, Falcon B, He S, Murzin AG, Murshudov G, Garringer HJ, et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature. 2017;547(7662):185–90.
Glynn C, Chun JE, Donahue CC, Nadler MJS, Fan Z, Hyman BT. Reconstitution of the Alzheimer’s disease tau core structure from recombinant tau(297–391) yields variable quaternary structures as seen by negative stain and cryo-EM. Biochemistry. 2024;63(2):194–201.
Carlomagno Y, Manne S, DeTure M, Prudencio M, Zhang YJ, Hanna Al-Shaikh R, et al. The AD tau core spontaneously self-assembles and recruits full-length tau to filaments. Cell Rep. 2021;34(11): 108843.
Cisek K, Cooper GL, Huseby CJ, Kuret J. Structure and mechanism of action of tau aggregation inhibitors. Curr Alzheimer Res. 2014;11(10):918–27.
Hosokawa M, Arai T, Masuda-Suzukake M, Nonaka T, Yamashita M, Akiyama H, et al. Methylene blue reduced abnormal tau accumulation in P301L tau transgenic mice. PLoS ONE. 2012;7(12): e52389.
Panza F, Solfrizzi V, Seripa D, Imbimbo BP, Lozupone M, Santamato A, et al. Tau-centric targets and drugs in clinical development for the treatment of Alzheimer’s disease. Biomed Res Int. 2016;2016:3245935.
Crowe A, James MJ, Lee VM, Smith AB 3rd, Trojanowski JQ, Ballatore C, et al. Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation. J Biol Chem. 2013;288(16):11024–37.
Seo D-H, Huh YH, Cheong H-K, Kim E-H, Lim J-S, Lee MJ, et al. Mechanism of methylene blue inducing the disulfide bond formation of tubulin-associated unit proteins. JACS Au. 2024;4(7):2451–5.
Rane JS, Bhaumik P, Panda D. Curcumin inhibits tau aggregation and disintegrates preformed tau filaments in vitro. J Alzheimers Dis. 2017;60(3):999–1014.
Zhong H, Liu H, Liu H. Molecular mechanism of tau misfolding and aggregation: insights from molecular dynamics simulation. Curr Med Chem. 2024;31(20):2855–71.
Necula M, Chirita CN, Kuret J. Cyanine dye N744 inhibits tau fibrillization by blocking filament extension: implications for the treatment of tauopathic neurodegenerative diseases. Biochemistry. 2005;44(30):10227–37.
Landau M, Sawaya MR, Faull KF, Laganowsky A, Jiang L, Sievers SA, et al. Towards a pharmacophore for amyloid. PLoS Biol. 2011;9(6): e1001080.
Griner SL, Seidler P, Bowler J, Murray KA, Yang TP, Sahay S, et al. Structure-based inhibitors of amyloid beta core suggest a common interface with tau. eLife. 2019;8:e46924.
Subedi S, Sasidharan S, Nag N, Saudagar P, Tripathi T. Amyloid cross-seeding: mechanism, implication, and inhibition. Molecules. 2022;27(6):1776.
Huang F, Wang J, Qu A, Shen L, Liu J, Liu J, et al. Maintenance of amyloid beta peptide homeostasis by artificial chaperones based on mixed-shell polymeric micelles. Angew Chem Int Ed Engl. 2014;53(34):8985–90.
Dhami KB, Karki S, Parks A, Nichols CG, Nichols MR. Development of beta-sheet structure in Abeta aggregation intermediates diminishes exposed hydrophobic surface area and enhances proinflammatory activity. Biochim Biophys Acta Proteins Proteom. 2022;1870(9): 140817.
Qi R, Luo Y, Wei G, Nussinov R, Ma B. Aβ, “stretching-and-packing” cross-seeding mechanism can trigger tau protein aggregation. J Phys Chem Lett. 2015;6(16):3276–82.
Griner SL, Seidler P, Bowler J, Murray KA, Yang TP, Sahay S, et al. Structure-based inhibitors of amyloid beta core suggest a common interface with tau. Elife. 2019;8:46924.
Fernandez-Ramirez MDC, Ng KK, Menendez M, Laurents DV, Hervas R, Carrion-Vazquez M. Expanded conformations of monomeric tau initiate its amyloidogenesis. Angew Chem Int Ed Engl. 2023;62(19): e202209252.
Kumar D, Kumar P. An in-silico investigation of key lysine residues and their selection for clearing off abeta and holo-AbetaPP through ubiquitination. Interdiscip Sci. 2019;11(4):584–96.
Miller Y, Ma B, Nussinov R. Synergistic interactions between repeats in tau protein and Abeta amyloids may be responsible for accelerated aggregation via polymorphic states. Biochemistry. 2011;50(23):5172–81.
Raz Y, Miller Y. Interactions between Abeta and mutated Tau lead to polymorphism and induce aggregation of Abeta-mutated tau oligomeric complexes. PLoS ONE. 2013;8(8): e73303.
Vasconcelos B, Stancu IC, Buist A, Bird M, Wang P, Vanoosthuyse A, et al. Heterotypic seeding of Tau fibrillization by pre-aggregated Abeta provides potent seeds for prion-like seeding and propagation of Tau-pathology in vivo. Acta Neuropathol. 2016;131(4):549–69.
Gotz J, Chen F, van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science. 2001;293(5534):1491–5.
Du Z, Li M, Ren J, Qu X. Current strategies for modulating abeta aggregation with multifunctional agents. Acc Chem Res. 2021;54(9):2172–84.
Di Natale G, Sabatino G, Sciacca MFM, Tosto R, Milardi D, Pappalardo G. Aβ and Tau interact with metal ions, lipid membranes and peptide-based amyloid inhibitors: are these common features relevant in Alzheimer’s disease? Molecules. 2022;27(16):5066.
Mancini RS, Wang Y, Weaver DF. Phenylindanes in brewed coffee inhibit amyloid-beta and tau aggregation. Front Neurosci. 2018;12:735.
Dolai S, Shi W, Corbo C, Sun C, Averick S, Obeysekera D, et al. “Clicked” sugar-curcumin conjugate: modulator of amyloid-beta and tau peptide aggregation at ultralow concentrations. ACS Chem Neurosci. 2011;2(12):694–9.
Ngo ST, Li MS. Curcumin binds to Abeta1-40 peptides and fibrils stronger than ibuprofen and naproxen. J Phys Chem B. 2012;116(34):10165–75.
Zhao LN, Chiu SW, Benoit J, Chew LY, Mu Y. The effect of curcumin on the stability of Abeta dimers. J Phys Chem B. 2012;116(25):7428–35.
Yang H, Zeng F, Luo Y, Zheng C, Ran C, Yang J. Curcumin scaffold as a multifunctional tool for Alzheimer’s disease research. Molecules. 2022;27(12).
Gao C, Wang Y, Sun J, Han Y, Gong W, Li Y, et al. Neuronal mitochondria-targeted delivery of curcumin by biomimetic engineered nanosystems in Alzheimer’s disease mice. Acta Biomater. 2020;108:285–99.
Pasieka A, Panek D, Szalaj N, Espargaro A, Wieckowska A, Malawska B, et al. Dual inhibitors of amyloid-beta and tau aggregation with amyloid-beta disaggregating properties: extended in cellulo, in silico, and kinetic studies of multifunctional anti-Alzheimer’s agents. ACS Chem Neurosci. 2021;12(11):2057–68.
Ramesh M, Acharya A, Murugan NA, Ila H, Govindaraju T. Thiophene-based dual modulators of Abeta and Tau aggregation. ChemBioChem. 2021;22(23):3348–57.
Park S, Shin J, Kim K, Kim D, Lee WS, Lee J, et al. Modulation of amyloid and tau aggregation to alleviate cognitive impairment in a transgenic mouse model of Alzheimer’s disease. ACS Pharmacol Transl Sci. 2024;7(9):2650–61.
Jakubowski JM, Orr AA, Le DA, Tamamis P. Correction to “interactions between curcumin derivatives and amyloid-β fibrils: insights from molecular dynamics simulations.” J Chem Inf Model. 2020;60(8):4108.
Zhan C, Chen Y, Tang Y, Wei G. Green tea extracts EGCG and EGC display distinct mechanisms in disrupting Abeta(42) protofibril. ACS Chem Neurosci. 2020;11(12):1841–51.
Wobst HJ, Sharma A, Diamond MI, Wanker EE, Bieschke J. The green tea polyphenol (-)-epigallocatechin gallate prevents the aggregation of tau protein into toxic oligomers at substoichiometric ratios. FEBS Lett. 2015;589(1):77–83.
Seidler PM, Murray KA, Boyer DR, Ge P, Sawaya MR, Hu CJ, et al. Structure-based discovery of small molecules that disaggregate Alzheimer’s disease tissue derived tau fibrils in vitro. Nat Commun. 2022;13(1):5451.
Zhan C, Chen Y, Tang Y, Wei G. Green tea extracts EGCG and EGC display distinct mechanisms in disrupting Aβ42 protofibril. ACS Chem Neurosci. 2020;11(12):1841–51.
Meng F, Bellaiche MMJ, Kim JY, Zerze GH, Best RB, Chung HS. Highly disordered amyloid-beta monomer probed by single-molecule FRET and MD simulation. Biophys J. 2018;114(4):870–84.
Levine ZA, Larini L, LaPointe NE, Feinstein SC, Shea JE. Regulation and aggregation of intrinsically disordered peptides. Proc Natl Acad Sci U S A. 2015;112(9):2758–63.
Im D, Heo CE, Son MK, Park CR, Kim HI, Choi JM. Kinetic modulation of amyloid-beta (1–42) aggregation and toxicity by structure-based rational design. J Am Chem Soc. 2022;144(4):1603–11.
Chung YJ, Lee BI, Park CB. Multifunctional carbon dots as a therapeutic nanoagent for modulating Cu(ii)-mediated beta-amyloid aggregation. Nanoscale. 2019;11(13):6297–306.
Zhou Y, Kandel N, Bartoli M, Serafim LF, ElMetwally AE, Falkenberg SM, et al. Structure-activity relationship of carbon nitride dots in inhibiting tau aggregation. Carbon N Y. 2022;193:1–16.
Zhang W, Smith N, Zhou Y, McGee CM, Bartoli M, Fu S, et al. Carbon dots as dual inhibitors of tau and amyloid-beta aggregation for the treatment of Alzheimer’s disease. Acta Biomater. 2024;183:341–55.
Sinha S, Lopes DH, Du Z, Pang ES, Shanmugam A, Lomakin A, et al. Lysine-specific molecular tweezers are broad-spectrum inhibitors of assembly and toxicity of amyloid proteins. J Am Chem Soc. 2011;133(42):16958–69.
Di J, Siddique I, Li Z, Malki G, Hornung S, Dutta S, et al. The molecular tweezer CLR01 improves behavioral deficits and reduces tau pathology in P301S-tau transgenic mice. Alzheimers Res Ther. 2021;13(1):6.
Lu J, Cao Q, Wang C, Zheng J, Luo F, Xie J, et al. Structure-based peptide inhibitor design of amyloid-beta aggregation. Front Mol Neurosci. 2019;12:54.
Sievers SA, Karanicolas J, Chang HW, Zhao A, Jiang L, Zirafi O, et al. Structure-based design of non-natural amino-acid inhibitors of amyloid fibril formation. Nature. 2011;475(7354):96–100.
Gandini A, Gonçalves AE, Strocchi S, Albertini C, Janočková J, Tramarin A, et al. Discovery of dual Aβ/Tau inhibitors and evaluation of their therapeutic effect on a drosophila model of Alzheimer’s disease. ACS Chem Neurosci. 2022;13(23):3314–29.
Haj E, Losev Y, Guru KrishnaKumar V, Pichinuk E, Engel H, Raveh A, et al. Integrating in vitro and in silico approaches to evaluate the “dual functionality” of palmatine chloride in inhibiting and disassembling Tau-derived VQIVYK peptide fibrils. Biochim Biophys Acta Gen Subj. 2018;1862(7):1565–75.
Hurtado DE, Molina-Porcel L, Iba M, Aboagye AK, Paul SM, Trojanowski JQ, et al. Abeta accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am J Pathol. 2010;177(4):1977–88.
Koller EJ, Ibanez KR, Vo Q, McFarland KN, Gonzalez De La Cruz E, Zobel L, et al. Combinatorial model of amyloid beta and tau reveals synergy between amyloid deposits and tangle formation. Neuropathol Appl Neurobiol. 2022;48(2):e12779.
Pascoal TA, Mathotaarachchi S, Mohades S, Benedet AL, Chung CO, Shin M, et al. Amyloid-beta and hyperphosphorylated tau synergy drives metabolic decline in preclinical Alzheimer’s disease. Mol Psychiatry. 2017;22(2):306–11.
Therriault J, Benedet AL, Pascoal TA, Mathotaarachchi S, Savard M, Chamoun M, et al. APOEepsilon4 potentiates the relationship between amyloid-beta and tau pathologies. Mol Psychiatry. 2021;26(10):5977–88.
Makhoba XH, Viegas C Jr, Mosa RA, Viegas FPD, Pooe OJ. Potential impact of the multi-target drug approach in the treatment of some complex diseases. Drug Des Devel Ther. 2020;14:3235–49.
Aksman LM, Oxtoby NP, Scelsi MA, Wijeratne PA, Young AL, Alves IL, et al. A data-driven study of Alzheimer’s disease related amyloid and tau pathology progression. Brain. 2023;146(12):4935–48.
Otaki H, Taguchi Y, Nishida N. Conformation-dependent influences of hydrophobic amino acids in two in-register parallel beta-sheet amyloids, an alpha-synuclein amyloid and a local structural model of PrP(Sc). ACS Omega. 2022;7(35):31271–88.
Oueslati A, Ximerakis M, Vekrellis K. Protein transmission, seeding and degradation: key steps for alpha-synuclein prion-like propagation. Exp Neurobiol. 2014;23(4):324–36.
Lindquist SL, Kelly JW. Chemical and biological approaches for adapting proteostasis to ameliorate protein misfolding and aggregation diseases-progress and prognosis. Cold Spring Harbor Persp Biol. 2011;3(12):a004507–a004507.
Jeon YH, Lee JY, Kim S. Chemical modulators working at pharmacological interface of target proteins. Bioorg Med Chem. 2012;20(6):1893–901.
Honig LS, Barakos J, Dhadda S, Kanekiyo M, Reyderman L, Irizarry M, et al. ARIA in patients treated with lecanemab (BAN2401) in a phase 2 study in early Alzheimer’s disease. Alzheimers Dement. 2023;9(1): e12377.
Agarwal A, Gupta V, Brahmbhatt P, Desai A, Vibhute P, Joseph-Mathurin N, et al. Amyloid-related imaging abnormalities in alzheimer disease treated with anti-amyloid-beta therapy. Radiographics. 2023;43(9): e230009.
Baek MS, Cho H, Lee HS, Lee JH, Ryu YH, Lyoo CH. Effect of APOE epsilon4 genotype on amyloid-beta and tau accumulation in Alzheimer’s disease. Alzheimers Res Ther. 2020;12(1):140.
Farfel JM, Yu L, De Jager PL, Schneider JA, Bennett DA. Association of APOE with tau-tangle pathology with and without beta-amyloid. Neurobiol Aging. 2016;37:19–25.
Neff RA, Wang M, Vatansever S, Guo L, Ming C, Wang Q, et al. Molecular subtyping of Alzheimer’s disease using RNA sequencing data reveals novel mechanisms and targets. Sci Adv. 2021;7(2):eabb5398.
Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14(4):535–62.
Schafer KN, Cisek K, Huseby CJ, Chang E, Kuret J. Structural determinants of Tau aggregation inhibitor potency. J Biol Chem. 2013;288(45):32599–611.
Choi M, Ryu J, Vu HD, Kim D, Youn YJ, Park MH, et al. Transferrin-conjugated melittin-loaded l-arginine-coated iron oxide nanoparticles for mitigating beta-amyloid pathology of the 5XFAD mouse brain. Int J Mol Sci. 2023;24(19):14954.
Clark AJ, Davis ME. Increased brain uptake of targeted nanoparticles by adding an acid-cleavable linkage between transferrin and the nanoparticle core. Proc Natl Acad Sci U S A. 2015;112(40):12486–91.
Tashima T. Smart strategies for therapeutic agent delivery into brain across the blood-brain barrier using receptor-mediated transcytosis. Chem Pharm Bull. 2020;68(4):316–25.
Abulrob A, Sprong H, Van Bergen P, Henegouwen E, Stanimirovic D. The blood–brain barrier transmigrating single domain antibody: mechanisms of transport and antigenic epitopes in human brain endothelial cells. J Neurochem. 2005;95(4):1201–14.
Zheng F, Pang Y, Li L, Pang Y, Zhang J, Wang X, et al. Applications of nanobodies in brain diseases. Front Immunol. 2022;13: 978513.
Zhou L, Kodidela S, Godse S, Thomas-Gooch S, Kumar A, Raji B, et al. Targeted drug delivery to the central nervous system using extracellular vesicles. Pharmaceuticals. 2022;15(3):358.
Dardet JP, Serrano N, András IE, Toborek M. Overcoming blood-brain barrier resistance: implications for extracellular vesicle-mediated drug brain delivery. Front Drug Delivery. 2022;2:855017.
Cummings J, Zhou Y, Lee G, Zhong K, Fonseca J, Cheng F. Alzheimer’s disease drug development pipeline: 2023. Alzheimers Dement. 2023;9(2): e12385.
Food and Drug Administration. FDA grants accelerated approval for alzheimer’s disease treatment. 2023. https://www.fda.gov/news-events/press-announcements/fda-grants-accelerated-approval-alzheimers-disease-treatment
Cavazzoni P. FDA’s decision to approve new treatment for Alzheimer’s disease. 2021. US Food and Drug Administration. https://www.fda.gov/drugs/our-perspective/fdas-decision-approve-new-treatment-alzheimers-disease
Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–6.
Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov. 2011;10(9):698–712.
Xu Z, Jia S, Wang W, Yuan Z, Jan Ravoo B, Guo DS. Heteromultivalent peptide recognition by co-assembly of cyclodextrin and calixarene amphiphiles enables inhibition of amyloid fibrillation. Nat Chem. 2019;11(1):86–93.
Geng H, Gao D, Wang Z, Liu X, Cao Z, Xing C. Strategies for inhibition and disaggregation of amyloid-β fibrillation. Chin J Chem. 2022;40(4):524–38.
Acknowledgements
Not applicable.
Funding
This review was funded by the Basic Science Research Program of the National Research Foundation of Korea (NRF), which is funded by the Ministry of Science, ICT & Future Planning (RS-2023-00240010 to M.M., RS-2024-00450135 to Y.N. and RS-2023-00212388 to S.K.), and was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (RS-2023-KH138733 to M.M.).
Author information
Authors and Affiliations
Contributions
Y.K., V.K., and S.J.S. wrote the manuscript. Y.K., V.K., J.W., and S.J.S. conducted the literature search and interpreted the data. S.K. and M.M. conceived the project, designed the review, arranged the results, and revised the manuscript. S.K. and M.M. accepted full responsibility for the finished work, had access to the data, and controlled the decision to publish. All authors have approved the final version of the manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no conflict of interest.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Nam, Y., Shin, S.J., Kumar, V. et al. Dual modulation of amyloid beta and tau aggregation and dissociation in Alzheimer’s disease: a comprehensive review of the characteristics and therapeutic strategies. Transl Neurodegener 14, 15 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40035-025-00479-4
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40035-025-00479-4