Abstract
Before the controversial approval of humanized monoclonal antibody lecanemab, which binds to the soluble amyloid‐β protofibrils, all the treatments available earlier, for Alzheimer's disease (AD) were symptomatic. The researchers are still struggling to find a breakthrough in AD therapeutic medicine, which is partially attributable to lack in understanding of the structural information associated with the intrinsically disordered proteins and amyloids. One of the major challenges in this area of research is to understand the structural diversity of intrinsically disordered proteins under in vitro conditions. Therefore, in this review, we have summarized the in vitro applications of biophysical methods, which are aimed to shed some light on the heterogeneity, pathogenicity, structures and mechanisms of the intrinsically disordered protein aggregates associated with proteinopathies including AD. This review will also rationalize some of the strategies in modulating disease‐relevant pathogenic protein entities by small molecules using structural biology approaches and biophysical characterization. We have also highlighted tools and techniques to simulate the in vivo conditions for native and cytotoxic tau/amyloids assemblies, urge new chemical approaches to replicate tau/amyloids assemblies similar to those in vivo conditions, in addition to designing novel potential drugs.
Keywords: Alzheimer's disease, amyloids, biophysical methods, drug development, immunotherapy
1. INTRODUCTION
Alzheimer's disease (AD) is considered as the most prevalent neurodegenerative disorder throughout the world. This progressive disease leads to dementia and cognitive impairment. It is the sixth leading cause of death in the United States. 1 , 2 AD is particularly prevalent in the elderly and an approximately over 50 million people suffer from this disease worldwide. 3 , 4 , 5 The late‐stage progression of AD worsens the patient's health and often leads to fatal consequences even when coupled with symptomatic treatment. 6 , 7 , 8 In α‐synucleinopathies, neuronal death and synaptic plasticity were characterized by abnormal accumulation of α‐synuclein aggregates. 9 Inflammation and synaptic dysfunction mechanistically interconnected in Parkinson's disease (PD), and supports the identification of reliable biomarkers for early diagnostics and development of therapies for PD. 9 , 10 , 11 This acts as a substantial burden on the economy, healthcare, and social systems. 1 , 12 The Food and Drugs Administration (FDA) approved AD drugs give symptomatic relief to the patients but they do not cure the underlying cause. 4 , 13 Most of these drugs were developed based on the cholinergic 14 and glutamatergic hypothesis, where they function to balance the disturbances in the brain neurotransmitters release and help to alleviate cognitive decline. 3 , 8 , 14 , 15 , 16 Additionally, monoclonal antibodies against different amyloid‐β (Aβ), based on the amyloid hypothesis, moderately slows down the cognitive (mild) decline and reduces Aβ plaques, in patients with early stage of AD. 17 , 18 Therefore, there is a surge in demand for developing potential cost‐effective drugs and therapeutic approaches for the treatment of AD. These drugs should be able to interfere with or stop the pathways of heterogeneous pathological toxic oligomer formation and bottom‐line mechanisms and kinetics of AD or other proteinopathies based on the amyloid. 19 Development in the field of structural biology techniques provide detailed knowledge about the exact tau structures and conformations, populations, sizes, toxicity pathways, and posttranslational modifications. 20 , 21 , 22 , 23 , 24 Furthermore, knowledge of protein–protein interactions, protein binding sites, kinetics of folding and aggregation, transient and native secondary, tertiary and quaternary structures, Aβ conformations found in vivo or in vitro is demanding. 25 , 26 , 27 These goals can be achieved by the application of biophysical methods and imaging techniques particularly nuclear magnetic resonance (NMR), 28 , 29 , 30 , 31 , 32 , 33 dynamic light scattering, 34 fluorescence resonance energy transfer (FRET) biosensors, 35 , 36 , 37 cryogenic electron microscopy (cryo‐EM), 38 , 39 , 40 , 41 , 42 x‐ray crystallography, 43 , 44 , 45 transmission EM (TEM), 46 and atomic force microscopy (AFM) 47 , 48 all of which will be addressed in this review (Figure 1). This review will also summarize the updates for small molecule inhibitors for amyloids associated with AD, PD 49 , 50 and will correlate the use of the essential methods for studying protein–protein interactions, and binding affinity of protein‐drugs 51 or ligands such as surface plasmon resonance (SPR), 52 bio‐layer interferometry (BLI), 51 , 53 isothermal titration calorimetry (ITC), 54 , 55 and supported by molecular docking 56 , 57 , 58 , 59 , 60 , 61 (Table 1). Finally, all these techniques discussed here will provide the awaited structural information about the heterogeneous toxic protein aggregates in AD, 62 allow to mimic cytotoxic species in vivo or patient‐derived material, 23 , 63 accelerating the designing of small molecules and production of more effective disease‐modifying drugs and therapeutics.
FIGURE 1.

Schematic diagram showing the fate of amyloid β plaque and tau monomers surrounded by—a ring of structural and biophysical techniques used for the characterization of monomers, oligomers, and fibrils (first circle). Second layer of techniques (in square) are applied for high throughput screening of chemical library and measurement of affinity with target proteins. Outermost layer depicts structural characterization and affinity measurements outcomes leading to toxic clearance via immunotherapy, and gene therapy.
TABLE 1.
Structural characterization of proteins and ligands using common structural and biophysical techniques.
| Methods | Techniques | Properties | References |
|---|---|---|---|
| In vitro |
Fluorescence probes—ThT Congo red and ANS |
Quantification of amyloids | 93, 94, 95, 96 |
| CD and FTIR | Secondary structure | 97, 98, 99, 100 | |
| ITC | Binding affinity and stoichiometry | 101, 102, 103 | |
| NMR | Structures, interactions, and dynamics | 104, 105, 106 | |
| X‐ray crystallography | Atomic structures | 107, 108, 109, 110 | |
| Cryo‐EM | Atomic structures | 109, 111, 112, 113, 114, 115, 116 | |
| SAXS | Structure | 117 | |
| TEM, AFM | Quaternary structure and surface feature | 118 | |
| In vivo | Fluorescent reporter assay | Extent of folding in cell | 119 |
Abbreviations: AFM, atomic force microscopy; cryo‐EM, cryogenic electron microscopy; ITC, isothermal titration calorimetry; NMR, nuclear magnetic resonance; TEM, transmission electron microscopy; ThT, thioflavin T; SAXS, small‐angle x‐ray scattering.
2. PROTEIN AGGREGATION AND NEURODEGENERATIVE DISEASE
Aβ is produced by the proteolytic action of secretases (β and γ) on amyloid precursor protein (APP). 64 , 65 Aβ accumulation particularly Aβ42 is the predominant protein species found in the senile plaque of AD patients' brains. 66 , 67 , 68 , 69 Several kinetic interrogations have revealed that once Aβ42 monomers concentration achieves critical point, it leads to the assembly of soluble oligomers. 70 , 71 , 72 Aβ42 oligomers are neurotoxic 73 as well as they stimulate tau pathogenicity. 74 Therefore, development of therapeutic strategies focuses on reducing the toxic form of Aβ42, either by targeting secretase, amyloid clearance, or stabilizing the monomeric form. 73 , 75 , 76 , 77 , 78 Aβ lacks well defined structure (intrinsically disordered) and exists as heterogeneous assembly of distinct conformations. 79 Targeting intrinsically disordered proteins (IDP; e.g., Aβ, tau, α‐syn) will lead in part to the failure of conventional drugs development which follow lock and key interaction hypothesis. 80 , 81 Search for novel therapeutics against AD and other neurodegenerative disorders require information about interaction and mechanism of action of therapeutics to alter or clear the toxic forms of tau/Aβ. 79 , 81 For the screening and characterization of the therapeutics, various biophysical, computational, and mathematical modeling techniques, are frequently used. 82 Researchers working in this particular aspect are keenly interested to find drug candidates which specifically act by binding and stabilizing the IDP monomer 83 , 84 and thus significantly reduce the nucleation pathways. Successful stabilization of the monomeric IDP (tau/Aβ) can be achieved by interaction with the small molecules that act as aggregation inhibitors. The small molecule aggregation inhibitors can be screened and developed on the basis of the critical domains responsible for oligomerization of these IDPs. 82 , 85
Tau is a microtubule (MT)‐associated protein that is distributed in the axonal region of the neurons of healthy people but is sorted in the somatodendritic compartment in tauopathy patients including AD. 86 , 87 Tau plays a role in regulating enzymatic activity of kinases and phosphatases through localization and signal transduction mechanisms. 88 , 89 Mutations in tau protein are associated with conformational changes, changes in isoform composition, changed affinity to the MTs, altered posttranslational modifications and increased aggregation propensity. 90 AD is closely associated with pathologic tau modification such as phosphorylation of specific residues and oligomerization. 91 Although tau toxicity is mainly enhanced by posttranslational modification and the mechanism behind this still remains unclear. 92
3. STRUCTURAL CHARACTERIZATION OF DISORDERED PROTEINS AND SMALL MOLECULES AS DRUG
3.1. X‐ray crystallography
Atomic level structure information of drug targets, designing, and optimization of lead molecules makes structural biology a powerful tool for drug discovery in human healthcare. 107 , 108 Structure determination of proteins and molecular complexes are limited to size of around 10–150 kDa as bulkier proteins, including membrane proteins, therefore it becomes difficult to crystallize and hence only crystal structures of few large size (greater than 150 kD) proteins are available. 109 , 110 Complete structures of protein (with high resolution) are required for structure‐based drug design. X‐ray crystallography provides for most of the protein structures needed for drug target except complex and membrane proteins which are difficult to crystallize. This drawback of x‐ray crystallography is now compensated by advances in cryo‐EM technique. 113 , 120 , 121
Proteins such as Aβ, tau, and α‐synuclein are the common IDPs; therefore, crystallization and structural determination of the monomeric forms of these proteins is extremely challenging. 21 , 64 , 122 Three‐dimensional structures determined by x‐ray crystallography provide more detailed information on the structure of Aβ. Recent updates of x‐ray crystallographic structural investigations of amyloids are showing that β‐sheet dimers of a β‐Hairpin derived from Aβ16–36 reorganize to tetrameric forms, which could be used as in vitro and in vivo models for full length brain derived Aβ for further investigations including drug discovery. 123 , 124 , 125 These crystal structures are aiding search for better small molecules in the form of novel chemical compounds, monoclonal antibodies and peptidomimetic construct that target amyloid aggregates. 125
3.2. Cryo‐EM
Presently, structural information about large macromolecules (>100 kD) and amyloids are primarily determined by cryo‐EM up to 2 Å resolution. 126 This can be achieved even with small molecules due to the advancements in cryo‐EM technology. 109 The advantages of simple and time resolved cryo‐EM over x‐ray diffraction technique are time saving for crystallization, smaller sample requirement, can cope with noncrystalline, higher size limit and ability to capture multiple highly dynamic protein structural conformations in solution. 111 , 112 These advantages of cryo‐EM have facilitated the development of drug targeting large and membrane protein family which is not possible through x‐ray crystallography. 113 , 114 , 115 , 116 Cryo‐EM is also contributing to the fragment‐based drug development as demonstrated by Saur et al., by his pilot study of β‐galactosidase and pyruvate kinase isozyme M2. 127
Nanoparticle labeled amyloid fibrils can be used for direct detection of amyloid fibrils from ex vivo sample and rapid profiling of polymorphism using cryo‐EM. 128 Cryo‐EM structure of brain derived Aβ fibrils are found to be right‐handed twisted polymorphic form but similar to protofilaments. 129 The mechanism of amyloid filament formation is disclosed and explained by the available high‐throughput Cryo‐EM structure of 76 different filaments. 23 Disordered proteins self‐assemble to form diverse higher order aggregates as well as amyloid fibrils. 119 Initially, large insoluble fibrils were known to be the primary factors associated for neurotoxicity in disease such as AD. 130 Recently, by the development of advance in vitro and in vivo techniques, it has demonstrated that smaller prefibrillar aggregates are the major toxic species in many neurodegenerative diseases. 131 , 132 , 133 , 134 Cryo‐EM structure of paired helical filament (PHF) and straight filament associated with AD, 63 , 135 Pick's disease, 136 corticobasal degeneration, 137 and chronic traumatic encephalopathy 138 has been determined. Cryo‐EM structure of in vitro tau filament assembly provides an optimistic model system to investigate the formation of different tau folds that define distinct tauopathies. The recent attainment for reporting fabrication of recombinant tau (297–391) into filaments identical to those of AD and chronic traumatic encephalopathy will play a crucial role in future investigations into the structural diversity of amyloids 23 and understanding the mechanism behind tauopathies. 38 Reweighted hierarchical chain growth (RHCG) algorithm method is a computationally common efficient alternative for MD where it combines pre‐sampled chain fragments in a statistically reproducible manner into ensembles of full‐length atomically detailed biomolecular structures. 139 RHCG was used to determine global structure of tau K18 protein by emerging from the local structure. RHCG integrates experimental data on local structure into the assembly process in a systematic manner and combines Bayesian ensemble refinement with importance sampling to end up with well‐defined structures. 21
Structural information about IDPs including tau conformations in solutions has been made by small‐angle x‐ray scattering. 117 Several studies investigated amyloid aggregation based on direct monitoring of oligomers, the slow processes of IDPs, protein dynamics, and intramolecular distance distributions using single molecule FRET (smFRET). 140 , 141 , 142 , 143 , 144 Also, more information about conformational distributions and secondary structures identification of intrinsically disordered and partially folded proteins is obtained by using NMR spectroscopy. 104 , 105 , 106
3.3. Isothermal titration calorimetry
ITC is an irreplaceable tool and is considered a gold standard for the estimation of thermodynamic parameters associated with protein–drug interactions and provides idea about molecular interaction mechanisms without any requirement for labeling. 145 , 146 Measurement of the affinity of ligand for the target protein is crucial for the selection of hit to lead optimization and hence is essential in the field of drug development. 101 , 102 , 103 Affinity parameters provide the driving force responsible for association of ligand, whereas the kinetic parameters of ligand–protein interactions provide residence time which measures the lifetime of the drug‐target complex, where both kinetic and thermodynamic profiling of compounds could be proposed as predictive tool for selectivity of compounds, and efficacy of drugs in vivo. 147 , 148 , 149 Novel therapeutic approaches acquire knowledge about protein–protein interactions between Aβ and its binding partners including tau and ApoE protein which has been possible by ITC. 150 , 151 , 152 The interaction of Aβ42 with Na,K‐ATPase and subsequent oligomerization leads to inhibition of the enzyme activity in brain of AD patients and contributes to AD pathogenesis. 54 The measured thermodynamic parameters for the formation of Na,K‐ATPase: Aβ42 complex at different conformations acquired by ITC are alike, which is in line with the data of MD. Similarity of all conformations of Na,K‐ATPase interaction interfaces with Aβ allowed to cross‐screen potential inhibitors for this interaction and find pharmaceutical compounds including ZINC153412538, ZINC153412622, and ZINC153412744 that can block it. 54
3.4. Surface plasmon resonance
SPR technique is based on immobilization of one binding partner with biosensor. Quantitative estimation of interactions between biomolecules using SPR and BLI is a highly reliable technique in comparison with other techniques including ELISA and ITC. This is because of the ability to work in real time as well as through a high throughput method. 52 , 153 Protein complexes associated with neuropathies such as Aβ, tau and their interacting partners, or ligands as lead molecules have been studied using SPR. Recent work reported that ApoE is binding laterally along the amyloid fibrils. 154 , 155 Immobilization procedures of monomeric or fibrillar form of amyloidogenic proteins on a sensor chip are different including covalent immobilization, high affinity capture binding, and hydrophobic attachment but can be used by slight modifications depending on the purposes of investigation to facilitate attachment and enhance signal. 156 , 157 , 158 , 159 SPR is a sensor based technique and could suffer from nonspecific binding which could be minimized and eliminated by careful experimental design, such as reducing the expected binding sites concentration. 160 Zheng et al. developed Au nanoparticle‐based dual aptamer‐based SPR biosensor to detect picomolar concentrations of Aβ40 oligomers and fibrils. 156
3.5. Biolayer interferometry
The fundamental importance of cellular function/dysfunction is associated with intra‐ and inter‐protein associations including various other biochemical reactions such as enzymatic reactions. Studies of multivalent interactions associated with protein functions and exploration in the field of novel therapeutics can be possible either by immobilization or fluorescence labeling based interactive modality techniques. 146 BLI is reliable for high affinity multimeric peptides as it is independent of concentration for immobilization and measurements in contrast to low affinity dimeric peptides working in concentration dependent manner and high signal to noise ration. 146 , 161 In a high throughput screening study employing BLI and other techniques for active small molecules from natural extract against Aβ42, potential amyloid inhibitors were identified including baicalein, polyporenic acid C, and other components which accelerates the development of therapeutics for AD. 51 , 82 , 162
3.6. NMR spectroscopy
As a significant outcome of recent advances and breakthroughs in NMR spectroscopy including instrumentation, isotope labeling, pulse sequences, 30 2D methods, 29 and solid‐state NMR probes; NMR has become a significant tool for studying dynamic protein complexes, IDPs structures (TDP‐43) and specific conformations which are hard to be obtained with other techniques. 163 , 164 NMR is a versatile tool where it encompasses solid‐state NMR at the atomic structure level, solution NMR for dynamic molecular structures and the in vivo biochemistry of proteins (hemoglobin, TDP‐43), 31 in addition to the membrane and cellular level for structural biology. 28 , 32 , 164 Recent NMR studies provided deeper insights into the protein's functional mechanisms, tau filaments specific structures, Aβ‐40 oligomers cross seeded by Aβ‐42 oligomers motif srtuctres. 165 , 166 Recent studies are indicating the ability of NMR for binding affinity of ligand‐proteins through the NMR coupling constants and chemical shifts changes, 167 aiding to depict oligomeric intermediates in an amyloid assembly. 168 Recently, solid‐state 19F‐NMR underlined the recognition of the binding sites for a positron emission tomography (PET) tracer to Aβ40. 169 Overall NMR is providing and revealing gap knowledge and current understanding of the neurodegenerative diseases causing structures and underlying mechanisms in addition to aiding to design aggregation inhibitors. 168
3.7. Characterization of amyloid deposits in living cells
In vivo time resolved monitoring of cellular and molecular events became accessible after the emergence of fluorescent protein that is sensitive toward cellular conditions as pH, stability, photoreactivity, and biosensors. 170 , 171 , 172 Real‐time and accurate information about cerebral Aβ deposition which is pathological hallmark of AD, and its noninvasive early diagnostics became possible only with fluorescence imaging, 173 , 174 , 175 PET, 169 and MRI. 176 Tao et al. developed ultrasensitive probe AH2, a derivative of Thioflavin T for the fluorescence‐based early detection and spatial distribution of Aβ deposition in mice brain as well as access the efficacy of antiaging medicines. 177 For conformational dynamics and biomolecular interaction at scale of 1–10 nm distance in steady‐state condition, smFRET has been extensively used based on transfer of energy between two chromophores. 35 Advantages of smFRET over other structural techniques includes high sensitivity, quality, structural transition measurement in equilibrium, high specificity to labeled domain of macromolecules, 178 cell organelles 179 even whole native cells. 180 , 181 For successful drug discovery landscape, it became demanding to have meticulous knowledge about disease relevant amyloids at early stages of proteinopathies, for better drug protein interactions which is achieved by applying in vivo FRET. Hippocampal neurons were reported to contain Aβ in the early stages of AD. MircoRNA‐125b(miR‐125b) can be a promising biomarker for the detection of early AD as it is associated with tau hyperphosphorylation using biosensor for miR‐125b. 182 , 183
4. AMYLOID POLYMORPHISM
Large numbers of evidence are now pointing out toward the amyloid polymorphism accountable for the occurrence of different strains of amyloids with varied toxicity and pathological spreading. 128 Structural polymorphism in amyloid forming protein is now recognized as common property. 184 , 185 , 186 Prion protein with abnormal β‐sheet rich form (PrPsc) is hallmark of prion disease deposited in brain by autocatalytic conversion of PrPc triggered by lipid and small RNA. 187 Molecular structure of self‐assembled amyloid fibrils and aggregates of monomeric proteins differing in structural composition are responsible for existence of multiple strains of prion diseases which causes variation in the disease toxicity. 188 , 189 , 190 , 191 In the case of AD and other amyloid disease, pattern of deposition of amyloid fibrils is responsible for generation of different strains. 192 Paravastu et al. describe the structural model of periodically twisted fibrils formed by Aβ‐40 by solid‐state NMR and TEM associated with AD also known as twisted pair morphology. 193 Tau and α‐synuclein are now being considered as prion like because of their inherent templating property for misfolding and aggregation of monomeric proteins and ability to form distinct stains. 194 Different downstream effects have been observed due to tau aggregates having conformational differences. 195 , 196 From the transsynaptic pattern of tau seed spreading, it has been proposed that tau protein also follow prion‐like mechanism of disease propagation. 197 , 198 On the basis of atomic structure of amyloid like fibers, three different models have been proposed and illustrated in Figure 2, 184 : (i) packing polymorphism, amyloid segment arranged in a pair of zipper connected by double headed arrow results in different structural and function properties; (ii) segmental polymorphism in which steric zipper spines are formed by two or more amyloid proteins; (iii) heterosteric zipper is formed by on‐identical beta sheets through interdigitation. Models derived from solid‐state NMR and cryo‐EM are best describing the heterotypic interactions between sheets. 184 , 199 , 200 , 201
FIGURE 2.

Three different models for the Tau amyloid polymorph: (A) Packing polymorphism (PDB: 2ON9 and 4NP8), (B) Segmental polymorphism (PDB: 2Y3J), and (C) Steric zipper (PDB: 3FVA) protofilament based on composition and pattern of amyloid segments.
5. PREVENTIVE MEASURE FOR AD PROGRESSION
Despite the enormous efforts and extensive research for finding novel therapeutics to prevent, halt or reverse the progression of AD, 202 still substantially cureless. 4 , 25 , 203 , 204 , 205 , 206 There are still no effective neurodegenerative diseases therapeutics that are able to halt or counteract the disease progression and impairment of mental activities but have short term symptoms of reliving from memory loss and thinking complications. 4 , 16 , 207 Most of these suggested medications and strategies ended with disappointments in early stages of development or in clinical trials. 6 , 7 , 8 , 17 , 19 , 205 , 208 , 209 , 210 , 211 , 212 , 213 Many previously reported reviews herein discussed thoroughly immunotherapy against tau and amyloids and related monoclonal antibodies as a future therapy. 8 , 13 , 17 , 19 , 39 , 204 , 205 , 209 , 211 , 212 , 213 , 214 , 215 , 216 Researchers faces many hurdles from two decades of failing attempts, unpleasant results for clinical trials of vaccine for the toxic amyloid and over many years of controversy and doubts regarding the efficacy, safety, tolerance of immunotherapy for neurodegenerative diseases. Finally some of these obstacles was relieved by the recent FDA approval for intravenous drug Aducanumab in June 2021 and the accelerated approval pathway for Lecanemab in January 2023. 5 Aducanumab (Aduhelm, BIIB037, humanized mAbs derived B cells selective to Aβ sponsored by Biogen, Neurimmune) lecanemab (BAN2401, mAb158, IgG1 mAb sponsored by BioArctic AB, Biogen, Eisai Co., Ltd.). 217 , 218 , 219 These two humanized monoclonal antibodies selectively bind with oligomeric and fibrillar states with signs of reduction and clearance of Aβ plaques and soluble Aβ protofibrils and as a result neuronal function and calcium uptake were restored in AD patient without significant toxicity. Lecanemab had tenfold stronger affinity with protofibrils than fibrils whereas Aducanumab preferred binding to fibrils over protofibrils. These results could explain some of the findings and outcomes in the clinical trials with regard to efficacy and adverse effects and type of mAb used. 17 The breakthrough for Aducanumab for clearance of Aβ plaques was achieved by its ability to cross brain–blood barrier (BBB), selective and preferred bounding to parenchymal Aβ over vascular Aβ aggregates. Intensified recruitment of f Iba‐1‐positive microglia associated with reduced potency of the glycosylated form of aducanumab that suggested that FcγR‐mediated microglial participation and phagocytosis contributed a major event in Aβ clearance. 220 The reported clearance ability of both Lecanemab and Aducanumab for Aβ plaques is bringing back the legacy of amyloid hypothesis of AD that was doubted and lapsed behind years ago as consequence of repeated setbacks in Aβ immunotherapy clinical trials. 49 , 221 , 222 The FDA approval initiated, sparked once again the monoclonal antibodies as potential promising topline forefront therapeutics and brought back the concept of Aβ and tau or combination of them as major immunotherapeutic targets for the prevention and/or treatment of AD. 212 , 223 Still the AD is irreversible and inevitable and the scenario of ending AD dilemma is not fulfilled, 224 where other paradigms concerned with therapeutics and research development pipelines are pursuing and focusing on the use of small molecules to tackle these neurodegenerative disorders. 3 , 5 , 39 , 47 , 50 , 215 , 225
5.1. Therapeutic interventions using small molecules as drugs
Recent and earlier significant scientific reports acknowledged the toxicity, heterogenicity, and seeding ability of tau oligomers for disease propagation in neurodegenerative diseases including AD. 226 , 227 , 228 Also, parallel studies reported the significance of Aβ oligomers (Aβ) 229 in neuropathogenesis and formation of plaques. Also considerations for the prion like α‐synuclein 230 in addition to other cascades theories. 231 , 232 , 233 Based on these streamlines that emanated from scientific research, spectroscopic and modeling data, innovative therapeutics for protein misfolding diseases including AD in the present time are targeting these disease pertinent inducers, through different routes and mechanisms including inhibition of oligomerization/aggregation, enhancing the recurrent of nonpathogenic species, and inhibition/alternation of soluble tau oligomers or nontoxic entities, 8 , 225 , 226 , 234 , 235 , 236 , 237 herein this review is highlighting some small molecules.
5.2. Reduction of Aβ production
To selectively prevent brain toxicity, and disordered brain functions due to Aβ oligomers and plaques accumulation, curative strategies were developed to decrease Aβ production by inhibition of the APP proteolytic enzymes including (the beta‐site APP‐cleaving enzyme 1) BACE1 and the β and γ‐secretase. 50 , 238 This strategy is important to tackle the protein misfolding disease from the very beginning. The γ‐secretase inhibitor Semagacestat (NCT00594568) was tested in phase 3 clinical trials in 2021 and terminated due several to causes including depletion of endogenous proteins, deterioration of motor functional capability, higher occurrence of malignant melanoma and cutaneous melanoma, increased infections in clinical trials patients acquiring drug as compared to placebo 238 , 239 and successive loss of associated cellular functions. 238 Another two BACE1 inhibitors were atabecestat (JNJ‐54861911) and E2609 which showed high tendency to decrease Aβ in the rodents' CSF, but were discounted in phase 2/3 clinical trials in 2021for safety issues and adverse effects including a loss of brain volume, a transient drop in white blood cells, and elevation of liver enzymes in some people. 215 , 240 Verubecestat (MK‐8931) and Umibecestat (CNP‐520) are other small‐molecule β‐secretase inhibitors which reached phase 3 clinical trials and discontinued in 2018 due to worsening cognitive functions. 241 , 242
5.3. Inhibiting protein aggregation
Anti‐aggregation agents are developed to exert their actions by forming different types of bonds between an inhibitor or small drug molecule and the backbone or side chain residues of the pathogenic conformation of related target protein that may influence one or all phases of the aggregation processes. 243 Some of these molecules can break off the formation of oligomer aggregates, and initiate the disaggregation of toxic oligomers into the native monomers consequently forestalling the buildup of oligomeric species. 25 Tau hyperphosphorylation and dismantling from MTs and consequently going through undesirable folding and accumulation to form PHFs and neurofibrillary tangles (NFTs) is substantially contributing to neurodegeneration disorders. 244 , 245 PHFs are the hallmark in tauopathies including AD 202 , 246 occurring from the abnormal aggregation of tau protein through nucleated assembly mechanism 247 ; and have double‐helical ribbon morphology. 248
To date, there are continued efforts on research pipelines aiming at the progress of cytotoxic aggregates and mechanisms and hindering the intracellular accumulation of tau aggregates considering the heterogenicity of the tau structures and isoforms in AD patients 24 in addition to Aβ or both. 50 , 249 , 250 Toxic tau oligomers impeding and modulation is still considered an effective promising drug development array for AD treatments. 47 , 251 , 252
Conclusively in this direction many preclinical studies were reported to explore many synthetic and natural products including polyphenol compounds such as, curcumin and its soluble bioavailable derivatives and analogs. 194 Some of the curcumin derivatives proved to reduce tau aggregation and could be used as potential tau PET tracers for early detection of tau oligomers. 47 Decreasing the population of brain derived toxic tau oligomers and enhancing the formation of higher order tau oligomers from small toxic oligomers through different interactions pathways, 194 ending up with modified structural conformations correlating with toxicity decline. 47 , 194 , 226 , 253 Raloxifene and R− (−)‐apomorphine indicated early their ability to maintain the p‐tau‐treated cells with inhibition pathway and showed consistent cryoprotection. In addition to their ability to penetrate the BBB and to maintain the cognitive functions, making them potential AD future treatments. 254 Natural product protein amyloid/tau inhibitors listing here resveratrol, epigallocatechin gallate (EGCG), morin, amentoflavone, bilobetin, sequoiaflavone, sotetsuflavone, podocarpuflavone, ginkgetin, isoginkgetin, sciadopitysin, 25 retinol, tannic acid, and caffeine have been shown to reduce aggregation proven by in vitro and cell‐based approaches. 238 , 243 Scyllo‐inositol, hopeahainol A, and rifampicin, in addition to other natural products tested on animals showed to be helpful for the recovery from the memory impairments and synaptic dysfunctions with alleviating the Aβ levels and senile plaque accumulations. 255
In vitro and preclinical research reported compounds like methylene blue with different formulations, N744, rhodamines, aminothienopyridazines, cinnamaldehyde, epicatechin oxidized form, crocin, VB‐008, and N‐(3‐chloro‐1,4‐dihydro‐1,4‐dioxo‐2‐naphthalenyl)‐l‐tryptophan that showed potential inhibition and targeting of tau oligomers or Aβ aggregation or combined action. 225 , 256 , 257 , 258 Still, the majority of these target compounds and drugs are not well developed or discontinued. 224 These disappointments in the scene of drug development and discovery could be attributable in part to lack in understanding of the structural information associated with the IDPs and amyloids. This includes cellular or in vivo pathways of disease relevant amyloid inhibition by small molecules, the presence of dynamic heterogeneous structural conformation in AD brain patients, triggering the cytotoxicity pathways and cell death progamming 24 ; in addition to the drug delivery issues. 50 , 225 , 259
The modified form of the methylthioninium (MT) moiety LMTX (TRx0237) which is an ongoing promising therapeutic is being investigated more often as an inhibitor for tau causing disease aggregation with failing attempts in phase 2/3 clinical trials due to low efficicy, 4 , 8 , 260 and slow cognitive or functional decline in subjects with mild to moderate AD (reported 2022). A new phase 3 clinical trial to acquire the safety and efficacy of LMTX (8 mg/day and 16 mg/day) in the treatment of AD patients compared to the placebo 50 , 225 , 252 , 260 , 261 is still under evaluation. A phase II clinical trials (NCT02033941) to investigate the effect of grape seed polyphenolic extract GSPE on AD tau aggregation are still going on. 262 The target drug NPT200‐11(C28H39N5O3) sponsored by neuropore therapies for the curative treatments of PD which had completed phase 1 clinical trial was reported to beat back the oligomerization, misfolding, buildup of α‐synuclein. It also lowers down the toxicity and pathology in cortex, reduction of astrogliosis, and improves brain motor function in preclinical studies 263 but did not exceed this stage as a proved drug or of label medication. In 2020, an 18‐month phase 2a double‐blind, placebo‐controlled study was initiated by UCB Biopharma SRL, for evaluation of the efficacy, tolerability, safety, and pharmacokinetics of oral UCB0599 (the R‐enantiomer of NPT200‐11) in study participants with early PD and is expected to completed in 2024. 206
Tramiprosate or Homotaurine (ALZ‐801) as a small therapeutic molecule that inhibits amyloid aggregation in the brain by binding selectively to Asp23, Aβ42, Lys16, and Lys28 showed in clinical trials for orally administrated drug to reduce in the AD tau and fibrillary amyloid aggregations. This was achieved by providing monomer stability in the brain with enhanced cholinergic functions and stable cognition performance; still it failed to comply with phase 3 clinical trials and terminated due to efficiency issues in 2009. 264 Few years later phase 3/4 clinical trials on AD apolipoprotein E carriers ApoE3 and ApoE4 (NCT04770220) based on differences in cortical thickness and positive results were initiated by Alzheon Inc. in 2019 running at 85 sites across the United States, Canada, and Europe, and still trials under investigation and expected to be completed in 2024. 73 , 264
5.4. Agents modulating tau posttranslational modifications
Several scientific works and publications reported the use of small molecule to tackle the tauopathies including AD by alternation and manipulation of the tau posttranslational modifications like hyperphosphorylation, truncation, glycosylation, and acetylation. Herein, the most important modification is hyperphosphorylation where the p‐tau is the predominant protein in most AD cases. It accounts for disruption of synaptic function, causing tau self‐assembly and intraneuronal accumulation of PHFs, aggregation in the form of NFTs and eventually leading to sever AD symptoms. Many phosphate activators like phosphoprotein phosphatase 2A (PP2A)‐related drugs were developed to reduce tau phosphorylation including the ionic compound sodium selenite. It was tested in vitro and in vivo to activate the PP2A, the sub catalytic enzymatic unit, reversing memory deficits and reducing tau phosphorylation in animal models of AD and showed in vivo selectivity toward p‐tau. 225 , 265 Randomized controlled trials of oral supplementation may potentially slow neurodegeneration in AD. 266 Other drugs were developed to work as kinase inhibitors targeting tau phosphorylation enzymes 225 such as saracatinib (AZD0530) which is a fyn inhibitor and tideglusib (NP031112) which is an inhibitor of glycogen synthase kinase 3β (GSK‐3β). 267 In animal studies, it was reported that tideglusib reduced a range of disease factors and symptoms, including tau phosphorylation, amyloid aggregation and deposition, neuron loss, and gliosis in mouse brain. Tideglusib showed some reduction in brain atrophy in a studied a subgroup; however, this trial was negative on the primary outcome. 268 All studies up to date for tideglusib were with limited success and did not exceed the phase II trials endpoints regarding mid and mild AD. 269
Blarcamesine (ANAVEX 2‐73) another (GSK3‐β) functioning as an agonist of sigma‐1 chaperone protein and developed for AD, PD, and related dementia is in the clinical trials for AD (phase 2/3), PD dementia (phase 2), PD endpoints (reported 2022) data outcomes indicated that it could be restoring neural cell homeostasis and enhancing neuroplasticity. Clinical trials indicated reduction in early and mild cognitive impairment 14 with association of AD and dementia in clinical trials patients. Blarcamesine is now in clinical trials to evaluate the effects of ANAVEX2‐73‐AD‐004 on safety and efficacy of daily treatment (NCT04314934) and expected to be completed in 2024. 270
5.5. Molecular docking‐molecular dynamics simulation for potential small molecular therapeutics
Recently, to overcome some of the hurdles in drugs discovery scenario in relation to many neurodegenerative diseases; computer‐based molecular dynamics (MD)‐molecular docking researchers to observe protein–drug interaction events exhibiting different methodologies. 271 , 272 , 273 The significance of these methods is to predict the atomic and molecular 3D functional structures of biomolecules at different disease‐associated stages, 274 , 275 fast effective identification of potential safe drugs, 276 relating structural disorders to cellular functions, 277 and envision binding modes, investigating interaction of tau and Aβ with cellular membranes and peptides insertions in model membrane, 278 , 279 reducing the use of animal models. 273 Many research groups are aiming to design innovative small molecules therapeutics, predict, simulate and screen synthetic and natural drugs 280 with optimum affinity for active site of target proteins and/or folding intermidiates. 280 , 281 , 282 , 283 Another way, some research groups actively employing network biology and polypharmacology (network pharmacology), for data integration and multitarget drug development. 282 , 284 , 285 , 286 This will be providing a library for potential multi‐target multifunction candidate drugs to tackle neurodegenerative diseases, proposing new AD treatment strategies, and an acceleration pathway for drug development. 280 , 281 , 282 , 286 Recent studies are showing that it is possible through the in vitro, machine learning and experimental and computational studies to predict novel inhibitors of aggregation (lead compounds) and acetylcholinesterase inhibitors. 287 , 288 Aβ42 monomer is in random coil conformation including either helices or β‐strand content whereas Aβ42 oligomers are in the form of amorphous aggregates containing few β‐sheets. 287 This outcome could be important for search of matching small molecules that prevent aggregation and toxicity. MD simulations of the antagonist ADH‐31 against Aβ42 aggregation resolved the inhibitory mechanism of ADH‐31 and demonstrated future potential use as protofibril destabilizer. Experimental and MD simulation studies of β‐casein displays chaperone like activity and inhibits Aβ40/Aβ42 aggregation and extracellular plaque formation, therefore can be used as novel drug for AD. 290 Kinetic modulation of Aβ42 aggregation based on biophysical approaches including x‐ray and in silico experiments is another recent addition to disclose structure based rational design of Aβ42 fibril inhibitor. 291 Murray et al. research group screened small molecule library using EGCG pharmacophore by molecular docking and found two novel compounds (CNS‐11 and CNS‐11g) and experimentally demonstrate the ability to disaggregate tau PHF and α‐synuclein fibrils. 292 MD simulation was performed to reveal the putative binding sites disaggregation mechanism of CNS‐11 and CNS‐11g in bound form and unbound form, 292 where these compounds could be potential specific targets for treatment of synucleinopathies. 292 Similar study disclosed the inhibition mechanisms of EGCG and Genistein on the AD amyloid‐beta 42 peptide conformational changes employing MD and molecular docking. 293 For visualizing the morphology of amyloidogenic protein aggregates, TEM 46 and AFM 47 , 48 are frequently used in vitro. Both these techniques provide quantitative and qualitative information at the nanometer scale for quaternary structure features including the length, width, and surface feature of aggregates. 294 , 295 , 296
6. CONCLUSION
With the intricate pathophysiology of the neurodegenerative diseases including AD and hindrance for drug development trajectories, application of computational methods with structural and biophysical techniques, can significantly contribute to developing small molecules targeting distinct protein aggregates associated with disease pathogenesis. Availability of well‐defined structures of biomolecules, kinetics, and their biochemistry accelerate the progress of drug discovery. Detection and revealing different pathogenic structural variants pose a new challenge to prepare and replicate them for further testing, which could be a critical step for drug development trajectories and effective treatments of AD. This review pinpoint about recent development in structural and biophysical techniques especially for studying amyloid formation and its dynamics, where such obtained data could be critical for revealing the mystery of the amyloids and its role in pathogenic processes.
AUTHOR CONTRIBUTIONS
Sharif Arar: Writing – original draft. Md. Anzarul Haque: Writing – original draft. Rakez Kayed: Conceptualization; validation; supervision; funding acquisition; writing – review and editing; writing – original draft.
PEER REVIEW
The peer review history for this article is available at https://www.webofscience.com/api/gateway/wos/peer‐review/10.1002/prot.26561.
ACKNOWLEDGMENTS
The authors thank the Kayed lab members for their support. This work was supported by National Institute of Health USA grants (R.K.): AG054025, RF1AG077484, AG072458, R01AG077253, AG060718, and the Mitchell Center for Neurodegenerative Diseases. Sharif Arar supported by the University of Jordan through scientific visit fellowship and the Fulbright Visiting Scholar Program (The Binational Fulbright Commission in Jordan) for the year 2022–2023.
Arar S, Haque MA, Kayed R. Protein aggregation and neurodegenerative disease: Structural outlook for the novel therapeutics. Proteins. 2025;93(8):1314‐1329. doi: 10.1002/prot.26561
Sharif Arar and Md Anzarul Haque contributed equally to this review.
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the corresponding author upon reasonable request.
REFERENCES
- 1. Skaria AP. The economic and societal burden of Alzheimer disease: managed care considerations. Am J Manag Care. 2022;28(10 Suppl):S188‐S196. [DOI] [PubMed] [Google Scholar]
- 2. James BD, Leurgans SE, Hebert LE, Scherr PA, Yaffe K, Bennett DA. Contribution of Alzheimer disease to mortality in the United States. Neurology. 2014;82(12):1045‐1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Cummings J, Lee G, Nahed P, et al. Alzheimer's disease drug development pipeline: 2022. Alzheimers Dement. 2022;8(1):e12295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Yiannopoulou KG, Papageorgiou SG. Current and future treatments in Alzheimer disease: an update. J Cent Nerv Syst Dis. 2020;12:1179573520907397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Jadhav S, Avila J, Scholl M, et al. A walk through tau therapeutic strategies. Acta Neuropathol Commun. 2019;7(1):22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Cummings J. New approaches to symptomatic treatments for Alzheimer's disease. Mol Neurodegener. 2021;16(1):2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Soeda Y, Takashima A. New insights into drug discovery targeting tau protein. Front Mol Neurosci. 2020;13:590896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Calabresi P, Mechelli A, Natale G, Volpicelli‐Daley L, Di Lazzaro G, Ghiglieri V. Alpha‐synuclein in Parkinson's disease and other synucleinopathies: from overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis. 2023;14(3):176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Zeighami Y, Fereshtehnejad SM, Dadar M, et al. A clinical‐anatomical signature of Parkinson's disease identified with partial least squares and magnetic resonance imaging. Neuroimage. 2019;190:69‐78. [DOI] [PubMed] [Google Scholar]
- 11. Grozdanov V, Bousset L, Hoffmeister M, et al. Increased immune activation by pathologic alpha‐synuclein in Parkinson's disease. Ann Neurol. 2019;86(4):593‐606. [DOI] [PubMed] [Google Scholar]
- 12. Kayed R, Jackson GR, Estes DM, Barrett AD. Alzheimers disease: review of emerging treatment role for intravenous immunoglobulins. J Cent Nerv Syst Dis. 2011;3:67‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Imbimbo BP, Balducci C, Ippati S, Watling M. Initial failures of anti‐tau antibodies in Alzheimer's disease are reminiscent of the amyloid‐beta story. Neural Regen Res. 2023;18(1):117‐118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Hampel H, Mesulam MM, Cuello AC, et al. The cholinergic system in the pathophysiology and treatment of Alzheimer's disease. Brain. 2018;141(7):1917‐1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Alhazmi HA, Albratty M. An update on the novel and approved drugs for Alzheimer disease. Saudi Pharm J. 2022;30(12):1755‐1764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Anand P, Singh B. A review on cholinesterase inhibitors for Alzheimer's disease. Arch Pharm Res. 2013;36(4):375‐399. [DOI] [PubMed] [Google Scholar]
- 17. Soderberg L, Johannesson M, Nygren P, et al. Lecanemab, aducanumab, and gantenerumab—binding profiles to different forms of amyloid‐beta might explain efficacy and side effects in clinical trials for Alzheimer's disease. Neurotherapeutics. 2023;20(1):195‐206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. van Dyck CH, Swanson CJ, Aisen P, et al. Lecanemab in early Alzheimer's disease. N Engl J Med. 2023;388:9‐21. [DOI] [PubMed] [Google Scholar]
- 19. Guo YLS, Zeng LH, Tan J. Tau‐targeting therapy in Alzheimer's disease: critical advances and future opportunities. Ageing Neur Dis. 2022;2(3):11‐29. [Google Scholar]
- 20. Zabik NL, Imhof MM, Martic‐Milne S. Structural evaluations of tau protein conformation: methodologies and approaches. Biochem Cell Biol. 2017;95(3):338‐349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Stelzl LS, Pietrek LM, Holla A, et al. Global structure of the intrinsically disordered protein tau emerges from its local structure. JACS Au. 2022;2(3):673‐686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Avila J, Jimenez JS, Sayas CL, et al. Tau structures. Front Aging Neurosci. 2016;8:262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Lovestam S, Koh FA, van Knippenberg B, et al. Assembly of recombinant tau into filaments identical to those of Alzheimer's disease and chronic traumatic encephalopathy. Elife. 2022;11:e76494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Limorenko G, Lashuel HA. Revisiting the grammar of tau aggregation and pathology formation: how new insights from brain pathology are shaping how we study and target tauopathies. Chem Soc Rev. 2022;51(2):513‐565. [DOI] [PubMed] [Google Scholar]
- 25. Kreiser RP, Wright AK, Block NR, et al. Therapeutic strategies to reduce the toxicity of misfolded protein oligomers. Int J Mol Sci. 2020;21(22):8651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Pignataro MF, Herrera MG, Dodero VI. Evaluation of peptide/protein self‐assembly and aggregation by spectroscopic methods. Molecules. 2020;25(20):4854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Stefani M. Biochemical and biophysical features of both oligomer/fibril and cell membrane in amyloid cytotoxicity. FEBS J. 2010;277(22):4602‐4613. [DOI] [PubMed] [Google Scholar]
- 28. Hu Y, Cheng K, He L, et al. NMR‐based methods for protein analysis. Anal Chem. 2021;93(4):1866‐1879. [DOI] [PubMed] [Google Scholar]
- 29. Kanelis V, Forman‐Kay JD, Kay LE. Multidimensional NMR methods for protein structure determination. IUBMB Life. 2001;52(6):291‐302. [DOI] [PubMed] [Google Scholar]
- 30. Kovermann M, Rogne P, Wolf‐Watz M. Protein dynamics and function from solution state NMR spectroscopy. Q Rev Biophys. 2016;49:e6. [DOI] [PubMed] [Google Scholar]
- 31. Laurents DV. AlphaFold 2 and NMR spectroscopy: partners to understand protein structure, dynamics and function. Front Mol Biosci. 2022;9:906437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Luchinat E, Banci L. In‐cell NMR: a topical review. IUCrJ. 2017;4(Pt 2):108‐118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Reif B, Ashbrook SE, Emsley L, Hong M. Solid‐state NMR spectroscopy. Nat Rev Methods Primers. 2021;1:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Stetefeld J, McKenna SA, Patel TR. Dynamic light scattering: a practical guide and applications in biomedical sciences. Biophys Rev. 2016;8(4):409‐427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Lerner E, Barth A, Hendrix J, et al. FRET‐based dynamic structural biology: challenges, perspectives and an appeal for open‐science practices. Elife. 2021;10:e60416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Maina KN, Smet‐Nocca C, Bitan G. Using FRET‐based biosensor cells to study the seeding activity of tau and alpha‐synuclein. Methods Mol Biol. 2023;2551:125‐145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Kim H, Seong J. Fluorescent protein‐based autophagy biosensors. Materials. 2021;14(11):3019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Scheres SH, Zhang W, Falcon B, Goedert M. Cryo‐EM structures of tau filaments. Curr Opin Struct Biol. 2020;64:17‐25. [DOI] [PubMed] [Google Scholar]
- 39. Seidler PM, Murray KA, Boyer DR, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Shi Y, Murzin AG, Falcon B, et al. Cryo‐EM structures of tau filaments from Alzheimer's disease with PET ligand APN‐1607. Acta Neuropathol. 2021;141(5):697‐708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Lippens G, Gigant B. Elucidating tau function and dysfunction in the era of cryo‐EM. J Biol Chem. 2019;294(24):9316‐9325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Mukherjee S, Dubois C, Perez K, et al. Quantitative proteomics of tau and Abeta in detergent fractions from Alzheimer's disease brains. J Neurochem. 2023;164(4):529‐552. [DOI] [PubMed] [Google Scholar]
- 43. Chourrout M, Roux M, Boisvert C, et al. Brain virtual histology with X‐ray phase‐contrast tomography part II: 3D morphologies of amyloid‐beta plaques in Alzheimer's disease models. Biomed Opt Express. 2022;13(3):1640‐1653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Figley MD, Gu W, Nanson JD, et al. SARM1 is a metabolic sensor activated by an increased NMN/NAD(+) ratio to trigger axon degeneration. Neuron. 2021;109(7):1118‐1136 e1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Sivakumar M, Saravanan K, Saravanan V, et al. Discovery of new potential triplet acting inhibitor for Alzheimer's disease via X‐ray crystallography, molecular docking and molecular dynamics. J Biomol Struct Dyn. 2020;38(7):1903‐1917. [DOI] [PubMed] [Google Scholar]
- 46. Martinez‐Miguel M, Tatkiewicz W, Kober M, et al. Methods for the characterization of protein aggregates. Methods Mol Biol. 2022;2406:479‐497. [DOI] [PubMed] [Google Scholar]
- 47. Lo Cascio F, Puangmalai N, Ellsworth A, et al. Toxic tau oligomers modulated by novel curcumin derivatives. Sci Rep. 2019;9(1):19011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Zhou L, Kurouski D. Structural characterization of individual alpha‐synuclein oligomers formed at different stages of protein aggregation by atomic force microscopy‐infrared spectroscopy. Anal Chem. 2020;92(10):6806‐6810. [DOI] [PubMed] [Google Scholar]
- 49. Bittar A, Bhatt N, Kayed R. Advances and considerations in AD tau‐targeted immunotherapy. Neurobiol Dis. 2020;134:104707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Yao W, Yang H, Yang J. Small‐molecule drugs development for Alzheimer's disease. Front Aging Neurosci. 2022;14:1019412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Quilodran‐Casas C, Silva VLS, Arcucci R, Heaney CE, Guo Y, Pain CC. Digital twins based on bidirectional LSTM and GAN for modelling the COVID‐19 pandemic. Neurocomputing. 2022;470:11‐28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Young KA, Mancera RL. Review: investigating the aggregation of amyloid beta with surface plasmon resonance: do different approaches yield different results? Anal Biochem. 2022;654:114828. [DOI] [PubMed] [Google Scholar]
- 53. Ziu I, Laryea ET, Alashkar F, Wu CG, Martic S. A dip‐and‐read optical aptasensor for detection of tau protein. Anal Bioanal Chem. 2020;412(5):1193‐1201. [DOI] [PubMed] [Google Scholar]
- 54. Adzhubei AA, Tolstova AP, Strelkova MA, Mitkevich VA, Petrushanko IY, Makarov AA. Interaction interface of Abeta(42) with human Na,K‐ATPase studied by MD and ITC and inhibitor screening by MD. Biomedicine. 2022;10: 1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Cotrina EY, Gimeno A, Llop J, et al. Calorimetric studies of binary and ternary molecular interactions between transthyretin, abeta peptides, and small‐molecule chaperones toward an alternative strategy for Alzheimer's disease drug discovery. J Med Chem. 2020;63(6):3205‐3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Hassan M, Shahzadi S, Seo SY, Alashwal H, Zaki N, Moustafa AA. Molecular docking and dynamic simulation of AZD3293 and solanezumab effects against BACE1 to treat Alzheimer's disease. Front Comput Neurosci. 2018;12:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Iqbal S, Potharaju R, Naveen S, Lokanath NK, Mohanakrishnan AK, Gunasekaran K. Design, crystal structure determination, molecular dynamic simulation and MMGBSA calculations of novel p38‐alpha MAPK inhibitors for combating Alzheimer's disease. J Biomol Struct Dyn. 2022;40(13):6114‐6127. [DOI] [PubMed] [Google Scholar]
- 58. Jarosinska OD, Rudiger SGD. Molecular strategies to target protein aggregation in Huntington's disease. Front Mol Biosci. 2021;8:769184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Lohr T, Kohlhoff K, Heller GT, Camilloni C, Vendruscolo M. A small molecule stabilizes the disordered native state of the Alzheimer's Abeta peptide. ACS Chem Nerosci. 2022;13(12):1738‐1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Nam KH. Molecular dynamics‐from small molecules to macromolecules. Int J Mol Sci. 2021;22(7):3761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Shan Y, Mysore VP, Leffler AE, Kim ET, Sagawa S, Shaw DE. How does a small molecule bind at a cryptic binding site? PLoS Comput Biol. 2022;18(3):e1009817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Gerson JE, Mudher A, Kayed R. Potential mechanisms and implications for the formation of tau oligomeric strains. Crit Rev Biochem Mol Biol. 2016;51(6):482‐496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Fitzpatrick AWP, Falcon B, He S, et al. Cryo‐EM structures of tau filaments from Alzheimer's disease. Nature. 2017;547(7662):185‐190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Chen GF, Xu TH, Yan Y, et al. Amyloid beta: structure, biology and structure‐based therapeutic development. Acta Pharmacol Sin. 2017;38(9):1205‐1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Devkota S, Williams TD, Wolfe MS. Familial Alzheimer's disease mutations in amyloid protein precursor alter proteolysis by gamma‐secretase to increase amyloid beta‐peptides of >/=45 residues. J Biol Chem. 2021;296:100281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353‐356. [DOI] [PubMed] [Google Scholar]
- 67. Lee JC, Kim SJ, Hong S, Kim Y. Diagnosis of Alzheimer's disease utilizing amyloid and tau as fluid biomarkers. Exp Mol Med. 2019;51(5):1‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Lue LF, Sabbagh MN, Chiu MJ, et al. Plasma levels of Abeta42 and tau identified probable Alzheimer's dementia: findings in two cohorts. Front Aging Neurosci. 2017;9:226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Gu L, Guo Z. Alzheimer's Abeta42 and Abeta40 peptides form interlaced amyloid fibrils. J Neurochem. 2013;126(3):305‐311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta‐peptide. Nat Rev Mol Cell Biol. 2007;8(2):101‐112. [DOI] [PubMed] [Google Scholar]
- 71. Benilova I, Karran E, De Strooper B. The toxic Abeta oligomer and Alzheimer's disease: an emperor in need of clothes. Nat Neurosci. 2012;15(3):349‐357. [DOI] [PubMed] [Google Scholar]
- 72. Upadhaya AR, Lungrin I, Yamaguchi H, Fandrich M, Thal DR. High‐molecular weight Abeta oligomers and protofibrils are the predominant Abeta species in the native soluble protein fraction of the AD brain. J Cell Mol Med. 2012;16(2):287‐295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Tolar M, Hey J, Power A, Abushakra S. Neurotoxic soluble amyloid oligomers drive Alzheimer's pathogenesis and represent a clinically validated target for slowing disease progression. Int J Mol Sci. 2021;22: 6355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Fyfe I. Influence of amyloid‐beta on tau spread in Alzheimer disease explained. Nat Rev Neurol. 2022;18(6):318. [DOI] [PubMed] [Google Scholar]
- 75. Knowles TP, Vendruscolo M, Dobson CM. The amyloid state and its association with protein misfolding diseases. Nat Rev Mol Cell Biol. 2014;15(6):384‐396. [DOI] [PubMed] [Google Scholar]
- 76. Arosio P, Michaels TC, Linse S, et al. Kinetic analysis reveals the diversity of microscopic mechanisms through which molecular chaperones suppress amyloid formation. Nat Commun. 2016;7:10948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Habchi J, Arosio P, Perni M, et al. An anticancer drug suppresses the primary nucleation reaction that initiates the production of the toxic Abeta42 aggregates linked with Alzheimer's disease. Sci Adv. 2016;2(2):e1501244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Aprile FA, Sormanni P, Perni M, et al. Selective targeting of primary and secondary nucleation pathways in Abeta42 aggregation using a rational antibody scanning method. Sci Adv. 2017;3(6):e1700488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Habchi J, Tompa P, Longhi S, Uversky VN. Introducing protein intrinsic disorder. Chem Rev. 2014;114(13):6561‐6588. [DOI] [PubMed] [Google Scholar]
- 80. Heller GT, Bonomi M, Vendruscolo M. Structural ensemble modulation upon small‐molecule binding to disordered proteins. J Mol Biol. 2018;430(16):2288‐2292. [DOI] [PubMed] [Google Scholar]
- 81. Heller GT, Sormanni P, Vendruscolo M. Targeting disordered proteins with small molecules using entropy. Trends Biochem Sci. 2015;40(9):491‐496. [DOI] [PubMed] [Google Scholar]
- 82. Heller GT, Aprile FA, Michaels TCT, et al. Small‐molecule sequestration of amyloid‐beta as a drug discovery strategy for Alzheimer's disease. Sci Adv. 2020;6:eabb5924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Follis AV, Hammoudeh DI, Wang H, Prochownik EV, Metallo SJ. Structural rationale for the coupled binding and unfolding of the c‐Myc oncoprotein by small molecules. Chem Biol. 2008;15(11):1149‐1155. [DOI] [PubMed] [Google Scholar]
- 84. Clausen DM, Guo J, Parise RA, et al. In vitro cytotoxicity and in vivo efficacy, pharmacokinetics, and metabolism of 10074‐G5, a novel small‐molecule inhibitor of c‐Myc/Max dimerization. J Pharmacol Exp Ther. 2010;335(3):715‐727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Im D, Kim S, Yoon G, 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):1065‐1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol. 2017;133(5):665‐704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Binder LI, Frankfurter A, Rebhun LI. The distribution of tau in the mammalian central nervous system. J Cell Biol. 1985;101(4):1371‐1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Kanaan NM, Pigino GF, Brady ST, Lazarov O, Binder LI, Morfini GA. Axonal degeneration in Alzheimer's disease: when signaling abnormalities meet the axonal transport system. Exp Neurol. 2013;246:44‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Mueller RL, Combs B, Alhadidy MM, Brady ST, Morfini GA, Kanaan NM. Tau: a signaling hub protein. Front Mol Neurosci. 2021;14:647054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Ghetti B, Oblak AL, Boeve BF, Johnson KA, Dickerson BC, Goedert M. Invited review: frontotemporal dementia caused by microtubule‐associated protein tau gene (MAPT) mutations: a chameleon for neuropathology and neuroimaging. Neuropathol Appl Neurobiol. 2015;41(1):24‐46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Combs B, Christensen KR, Richards C, et al. Frontotemporal lobar dementia mutant tau impairs axonal transport through a protein phosphatase 1gamma‐dependent mechanism. J Neurosci. 2021;41(45):9431‐9451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Nieznanska H, Boyko S, Dec R, Redowicz MJ, Dzwolak W, Nieznanski K. Neurotoxicity of oligomers of phosphorylated tau protein carrying tauopathy‐associated mutation is inhibited by prion protein. Biochim Biophys Acta Mol Basis Dis. 2021;1867(11):166209. [DOI] [PubMed] [Google Scholar]
- 93. Nilsson MR. Techniques to study amyloid fibril formation in vitro. Methods. 2004;34(1):151‐160. [DOI] [PubMed] [Google Scholar]
- 94. Maezawa I, Hong HS, Liu R, et al. Congo red and thioflavin‐T analogs detect Abeta oligomers. J Neurochem. 2008;104(2):457‐468. [DOI] [PubMed] [Google Scholar]
- 95. Reinke AA, Gestwicki JE. Insight into amyloid structure using chemical probes. Chem Biol Drug des. 2011;77(6):399‐411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Hawe A, Sutter M, Jiskoot W. Extrinsic fluorescent dyes as tools for protein characterization. Pharm Res. 2008;25(7):1487‐1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Bhak G, Lee JH, Hahn JS, Paik SR. Granular assembly of alpha‐synuclein leading to the accelerated amyloid fibril formation with shear stress. PloS One. 2009;4(1):e4177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Lee JH, Bhak G, Lee SG, Paik SR. Instantaneous amyloid fibril formation of alpha‐synuclein from the oligomeric granular structures in the presence of hexane. Biophys J. 2008;95(2):L16‐L18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Ku SH, Park CB. Highly accelerated self‐assembly and fibrillation of prion peptides on solid surfaces. Langmuir. 2008;24(24):13822‐13827. [DOI] [PubMed] [Google Scholar]
- 100. Ha C, Park CB. Template‐directed self‐assembly and growth of insulin amyloid fibrils. Biotechnol Bioeng. 2005;90(7):848‐855. [DOI] [PubMed] [Google Scholar]
- 101. Olsson TS, Williams MA, Pitt WR, Ladbury JE. The thermodynamics of protein‐ligand interaction and solvation: insights for ligand design. J Mol Biol. 2008;384(4):1002‐1017. [DOI] [PubMed] [Google Scholar]
- 102. Wang Y, Edalji RP, Panchal SC, Sun C, Djuric SW, Vasudevan A. Are we there yet? Applying thermodynamic and kinetic profiling on embryonic ectoderm development (EED) hit‐to‐lead program. J Med Chem. 2017;60(20):8321‐8335. [DOI] [PubMed] [Google Scholar]
- 103. Linkuviene V, Talibov VO, Danielson UH, Matulis D. Introduction of intrinsic kinetics of protein‐ligand interactions and their implications for drug design. J Med Chem. 2018;61(6):2292‐2302. [DOI] [PubMed] [Google Scholar]
- 104. Schwalbe M, Ozenne V, Bibow S, et al. Predictive atomic resolution descriptions of intrinsically disordered hTau40 and alpha‐synuclein in solution from NMR and small angle scattering. Structure. 2014;22(2):238‐249. [DOI] [PubMed] [Google Scholar]
- 105. Meier S, Blackledge M, Grzesiek S. Conformational distributions of unfolded polypeptides from novel NMR techniques. J Chem Phys. 2008;128(5):052204. [DOI] [PubMed] [Google Scholar]
- 106. Jensen MR, Salmon L, Nodet G, Blackledge M. Defining conformational ensembles of intrinsically disordered and partially folded proteins directly from chemical shifts. J Am Chem Soc. 2010;132(4):1270‐1272. [DOI] [PubMed] [Google Scholar]
- 107. Thomford NE, Senthebane DA, Rowe A, et al. Natural products for drug discovery in the 21st century: innovations for novel drug discovery. Int J Mol Sci. 2018;19(6):1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Zhu KF, Yuan C, Du YM, et al. Applications and prospects of cryo‐EM in drug discovery. Mil Med Res. 2023;10(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Wang HW, Wang JW. How cryo‐electron microscopy and X‐ray crystallography complement each other. Protein Sci. 2017;26(1):32‐39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Shi Y. A glimpse of structural biology through X‐ray crystallography. Cell. 2014;159(5):995‐1014. [DOI] [PubMed] [Google Scholar]
- 111. Bai XC, McMullan G, Scheres SH. How cryo‐EM is revolutionizing structural biology. Trends Biochem Sci. 2015;40(1):49‐57. [DOI] [PubMed] [Google Scholar]
- 112. Frank J. Time‐resolved cryo‐electron microscopy: recent progress. J Struct Biol. 2017;200(3):303‐306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Garcia‐Nafria J, Tate CG. Cryo‐electron microscopy: moving beyond X‐ray crystal structures for drug receptors and drug development. Annu Rev Pharmacol Toxicol. 2020;60:51‐71. [DOI] [PubMed] [Google Scholar]
- 114. Zhang X, Johnson RM, Drulyte I, et al. Evolving cryo‐EM structural approaches for GPCR drug discovery. Structure. 2021;29(9):963‐974 e966. [DOI] [PubMed] [Google Scholar]
- 115. Congreve M, de Graaf C, Swain NA, Tate CG. Impact of GPCR structures on drug discovery. Cell. 2020;181(1):81‐91. [DOI] [PubMed] [Google Scholar]
- 116. Liang YL, Khoshouei M, Radjainia M, et al. Phase‐plate cryo‐EM structure of a class B GPCR‐G‐protein complex. Nature. 2017;546(7656):118‐123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Mylonas E, Hascher A, Bernado P, Blackledge M, Mandelkow E, Svergun DI. Domain conformation of tau protein studied by solution small‐angle X‐ray scattering. Biochemistry. 2008;47(39):10345‐10353. [DOI] [PubMed] [Google Scholar]
- 118. Stine WB Jr, Dahlgren KN, Krafft GA, LaDu MJ. In vitro characterization of conditions for amyloid‐beta peptide oligomerization and fibrillogenesis. J Biol Chem. 2003;278(13):11612‐11622. [DOI] [PubMed] [Google Scholar]
- 119. Gregoire S, Irwin J, Kwon I. Techniques for monitoring protein misfolding and aggregation in vitro and in living cells. Korean J Chem Eng. 2012;29(6):693‐702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Garavito RM, Picot D, Loll PJ. Strategies for crystallizing membrane proteins. J Bioenerg Biomembr. 1996;28(1):13‐27. [PubMed] [Google Scholar]
- 121. Melnikov I, Svensson O, Bourenkov G, Leonard G, Popov A. The complex analysis of X‐ray mesh scans for macromolecular crystallography. Acta Crystallogr D Struct Biol. 2018;74(Pt 4):355‐365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Mignon J, Mottet D, Leyder T, Uversky VN, Perpete EA, Michaux C. Structural characterisation of amyloidogenic intrinsically disordered zinc finger protein isoforms DPF3b and DPF3a. Int J Biol Macromol. 2022;218:57‐71. [DOI] [PubMed] [Google Scholar]
- 123. Kreutzer AG, Samdin TD, Guaglianone G, Spencer RK, Nowick JS. X‐ray crystallography reveals parallel and antiparallel beta‐sheet dimers of a beta‐hairpin derived from Abeta(16‐36) that assemble to form different tetramers. ACS Chem Nerosci. 2020;11(15):2340‐2347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Yu L, Edalji R, Harlan JE, et al. Structural characterization of a soluble amyloid beta‐peptide oligomer. Biochemistry. 2009;48(9):1870‐1877. [DOI] [PubMed] [Google Scholar]
- 125. Streltsov VA, Varghese JN, Masters CL, Nuttall SD. Crystal structure of the amyloid‐beta p3 fragment provides a model for oligomer formation in Alzheimer's disease. J Neurosci. 2011;31(4):1419‐1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Yang Y, Zhang W, Murzin AG, et al. Cryo‐EM structures of amyloid‐beta filaments with the Arctic mutation (E22G) from human and mouse brains. Acta Neuropathol. 2023;145(3):325‐333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Saur M, Hartshorn MJ, Dong J, et al. Fragment‐based drug discovery using cryo‐EM. Drug Discov Today. 2020;25(3):485‐490. [DOI] [PubMed] [Google Scholar]
- 128. Cendrowska U, Silva PJ, Ait‐Bouziad N, et al. Unraveling the complexity of amyloid polymorphism using gold nanoparticles and cryo‐EM. Proc Natl Acad Sci U S A. 2020;117(12):6866‐6874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Kollmer M, Close W, Funk L, et al. Cryo‐EM structure and polymorphism of Abeta amyloid fibrils purified from Alzheimer's brain tissue. Nat Commun. 2019;10(1):4760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Hardy JA, Higgins GA. Alzheimer's disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184‐185. [DOI] [PubMed] [Google Scholar]
- 131. Vunnam N, Hansen S, Williams DC, et al. Fluorescence lifetime measurement of prefibrillar sickle hemoglobin oligomers as a platform for drug discovery in sickle cell disease. Biomacromolecules. 2022;23(9):3822‐3830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Banerjee S, Holcombe B, Ringold S, et al. Nanoscale infrared spectroscopy identifies structural heterogeneity in individual amyloid fibrils and prefibrillar aggregates. J Phys Chem B. 2022;126(31):5832‐5841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Guzzo A, Delarue P, Rojas A, Nicolai A, Maisuradze GG, Senet P. Wild‐type alpha‐synuclein and variants occur in different disordered dimers and pre‐fibrillar conformations in early stage of aggregation. Front Mol Biosci. 2022;9:910104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Yoshikawa Y, Yuzu K, Yamamoto N, et al. Pathway dependence of the formation and development of prefibrillar aggregates in insulin B chain. Molecules. 2022;27((13)):3964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Falcon B, Zhang W, Schweighauser M, et al. Tau filaments from multiple cases of sporadic and inherited Alzheimer's disease adopt a common fold. Acta Neuropathol. 2018;136(5):699‐708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Falcon B, Zhang W, Murzin AG, et al. Structures of filaments from Pick's disease reveal a novel tau protein fold. Nature. 2018;561(7721):137‐140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Zhang W, Tarutani A, Newell KL, et al. Novel tau filament fold in corticobasal degeneration. Nature. 2020;580(7802):283‐287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Falcon B, Zivanov J, Zhang W, et al. Novel tau filament fold in chronic traumatic encephalopathy encloses hydrophobic molecules. Nature. 2019;568(7752):420‐423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Pietrek LM, Stelzl LS, Hummer G. Structural ensembles of disordered proteins from hierarchical chain growth and simulation. Curr Opin Struct Biol. 2023;78:102501. [DOI] [PubMed] [Google Scholar]
- 140. Kalinin S, Peulen T, Sindbert S, et al. A toolkit and benchmark study for FRET‐restrained high‐precision structural modeling. Nat Methods. 2012;9(12):1218‐1225. [DOI] [PubMed] [Google Scholar]
- 141. Shammas SL, Garcia GA, Kumar S, et al. A mechanistic model of tau amyloid aggregation based on direct observation of oligomers. Nat Commun. 2015;6:7025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Schuler B, Soranno A, Hofmann H, Nettels D. Single‐molecule FRET spectroscopy and the polymer physics of unfolded and intrinsically disordered proteins. Annu Rev Biophys. 2016;45:207‐231. [DOI] [PubMed] [Google Scholar]
- 143. Borgia A, Borgia MB, Bugge K, et al. Extreme disorder in an ultrahigh‐affinity protein complex. Nature. 2018;555(7694):61‐66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Brucale M, Schuler B, Samori B. Single‐molecule studies of intrinsically disordered proteins. Chem Rev. 2014;114(6):3281‐3317. [DOI] [PubMed] [Google Scholar]
- 145. Ladbury JE, Chowdhry BZ. Sensing the heat: the application of isothermal titration calorimetry to thermodynamic studies of biomolecular interactions. Chem Biol. 1996;3(10):791‐801. [DOI] [PubMed] [Google Scholar]
- 146. Schulte C, Solda A, Spanig S, et al. Multivalent binding kinetics resolved by fluorescence proximity sensing. Commun Biol. 2022;5(1):1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Zhang R, Monsma F. Binding kinetics and mechanism of action: toward the discovery and development of better and best in class drugs. Expert Opin Drug Discov. 2010;5(11):1023‐1029. [DOI] [PubMed] [Google Scholar]
- 148. Swinney DC. Biochemical mechanisms of drug action: what does it take for success? Nat Rev Drug Discov. 2004;3(9):801‐808. [DOI] [PubMed] [Google Scholar]
- 149. Su H, Xu Y. Application of ITC‐based characterization of thermodynamic and kinetic association of ligands with proteins in drug design. Front Pharmacol. 2018;9:1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Liu S, Park S, Allington G, et al. Targeting apolipoprotein E/amyloid beta binding by peptoid CPO_Abeta17‐21 P ameliorates Alzheimer's disease related pathology and cognitive decline. Sci Rep. 2017;7(1):8009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Nasica‐Labouze J, Nguyen PH, Sterpone F, et al. Amyloid beta protein and Alzheimer's disease: when computer simulations complement experimental studies. Chem Rev. 2015;115(9):3518‐3563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Wallin C, Hiruma Y, Warmlander S, et al. The neuronal tau protein blocks in vitro fibrillation of the amyloid‐beta (Abeta) peptide at the oligomeric stage. J Am Chem Soc. 2018;140(26):8138‐8146. [DOI] [PubMed] [Google Scholar]
- 153. Lee TH, Hirst DJ, Kulkarni K, Del Borgo MP, Aguilar MI. Exploring molecular‐biomembrane interactions with surface plasmon resonance and dual polarization interferometry technology: expanding the spotlight onto biomembrane structure. Chem Rev. 2018;118(11):5392‐5487. [DOI] [PubMed] [Google Scholar]
- 154. Islam T, Gharibyan AL, Lee CC, Olofsson A. Morphological analysis of Apolipoprotein E binding to Abeta amyloid using a combination of surface Plasmon resonance, Immunogold labeling and scanning electron microscopy. BMC Biotechnol. 2019;19(1):97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Ziehm T, Buell AK, Willbold D. Role of hydrophobicity and charge of amyloid‐beta oligomer eliminating d‐peptides in the interaction with amyloid‐beta monomers. ACS Chem Nerosci. 2018;9(11):2679‐2688. [DOI] [PubMed] [Google Scholar]
- 156. Zheng Y, Geng X, Yang X, et al. Exploring interactions of aptamers with Abeta(40) amyloid aggregates and its application: detection of amyloid aggregates. Anal Chem. 2020;92(3):2853‐2858. [DOI] [PubMed] [Google Scholar]
- 157. Pillay K, Govender P. Novel insights into amylin aggregation. Biotechnol Biotechnol Equip. 2014;28(1):123‐135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Hibino E, Inoue R, Sugiyama M, Kuwahara J, Matsuzaki K, Hoshino M. Interaction between intrinsically disordered regions in transcription factors Sp1 and TAF4. Protein Sci. 2016;25(11):2006‐2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Lee YK, Lee KS, Kim WM, Sohn YS. Detection of amyloid‐beta42 using a waveguide‐coupled bimetallic surface plasmon resonance sensor chip in the intensity measurement mode. PloS One. 2014;9(6):e98992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Wittenberg NJ, Wootla B, Jordan LR, et al. Applications of SPR for the characterization of molecules important in the pathogenesis and treatment of neurodegenerative diseases. Expert Rev Neurother. 2014;14(4):449‐463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Ponzo I, Moller FM, Daub H, Matscheko N. A DNA‐based biosensor assay for the kinetic characterization of ion‐dependent aptamer folding and protein binding. Molecules. 2019;24(16):2877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Klein AN, Ziehm T, Tusche M, et al. Optimization of the all‐D peptide D3 for Abeta oligomer elimination. PloS One. 2016;11(4):e0153035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Huang C, Kalodimos CG. Structures of large protein complexes determined by nuclear magnetic resonance spectroscopy. Annu Rev Biophys. 2017;46:317‐336. [DOI] [PubMed] [Google Scholar]
- 164. Gronenborn AM, Polenova T. Introduction: biomolecular NMR spectroscopy. Chem Rev. 2022;122(10):9265‐9266. [DOI] [PubMed] [Google Scholar]
- 165. Kitoka K, Skrabana R, Gasparik N, Hritz J, Jaudzems K. NMR studies of tau protein in tauopathies. Front Mol Biosci. 2021;8:761227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Chang HW, Ma HI, Wu YS, et al. Site specific NMR characterization of abeta‐40 oligomers cross seeded by abeta‐42 oligomers. Chem Sci. 2022;13(29):8526‐8535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Gossert AD, Jahnke W. NMR in drug discovery: a practical guide to identification and validation of ligands interacting with biological macromolecules. Prog Nucl Magn Reson Spectrosc. 2016;97:82‐125. [DOI] [PubMed] [Google Scholar]
- 168. Karamanos TK, Jackson MP, Calabrese AN, et al. Structural mapping of oligomeric intermediates in an amyloid assembly pathway. Elife. 2019;8:e46574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Duan P, Chen KJ, Wijegunawardena G, et al. Binding sites of a positron emission tomography imaging agent in Alzheimer's beta‐amyloid fibrils studied using (19)F solid‐state NMR. J Am Chem Soc. 2022;144(3):1416‐1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Eason MG, Damry AM, Chica RA. Structure‐guided rational design of red fluorescent proteins: towards designer genetically‐encoded fluorophores. Curr Opin Struct Biol. 2017;45:91‐99. [DOI] [PubMed] [Google Scholar]
- 171. Kamiyama D, Sekine S, Barsi‐Rhyne B, et al. Versatile protein tagging in cells with split fluorescent protein. Nat Commun. 2016;7:11046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Nienhaus K, Nienhaus GU. Fluorescent proteins for live‐cell imaging with super‐resolution. Chem Soc Rev. 2014;43(4):1088‐1106. [DOI] [PubMed] [Google Scholar]
- 173. Shimogawa M, Petersson EJ. New strategies for fluorescently labeling proteins in the study of amyloids. Curr Opin Chem Biol. 2021;64:57‐66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Fu H, Cui M. Fluorescent imaging of amyloid‐beta deposits in brain: an overview of probe development and a highlight of the applications for in vivo imaging. Curr Med Chem. 2018;25(23):2736‐2759. [DOI] [PubMed] [Google Scholar]
- 175. Gyasi YI, Pang YP, Li XR, et al. Biological applications of near infrared fluorescence dye probes in monitoring Alzheimer's disease. Eur J Med Chem. 2020;187:111982. [DOI] [PubMed] [Google Scholar]
- 176. Huang P, Zhang M. Magnetic resonance imaging studies of neurodegenerative disease: from methods to translational research. Neurosci Bull. 2023;39(1):99‐112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Tao R, Wang N, Shen T, et al. High‐fidelity imaging of amyloid‐beta deposits with an ultrasensitive fluorescent probe facilitates the early diagnosis and treatment of Alzheimer's disease. Theranostics. 2022;12(6):2549‐2559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Kilic S, Felekyan S, Doroshenko O, et al. Single‐molecule FRET reveals multiscale chromatin dynamics modulated by HP1alpha. Nat Commun. 2018;9(1):235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Dunkle JA, Wang L, Feldman MB, et al. Structures of the bacterial ribosome in classical and hybrid states of tRNA binding. Science. 2011;332(6032):981‐984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Lu M, Ma X, Mothes W. Illuminating the virus life cycle with single‐molecule FRET imaging. Adv Virus Res. 2019;105:239‐273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Munro JB, Gorman J, Ma X, et al. Conformational dynamics of single HIV‐1 envelope trimers on the surface of native virions. Science. 2014;346(6210):759‐763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Gao Y, Wennmalm S, Winblad B, Schedin‐Weiss S, Tjernberg LO. Live cell FRET imaging reveals amyloid beta‐peptide oligomerization in hippocampal neurons. Int J Mol Sci. 2021;22(9):4530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Zhang Q, Yin B, Huang Y, et al. A dual "turn‐on" biosensor based on AIE effect and FRET for in situ detection of miR‐125b biomarker in early Alzheimer's disease. Biosens Bioelectron. 2023;230:115270. [DOI] [PubMed] [Google Scholar]
- 184. Eisenberg D, Jucker M. The amyloid state of proteins in human diseases. Cell. 2012;148(6):1188‐1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Tanaka M, Collins SR, Toyama BH, Weissman JS. The physical basis of how prion conformations determine strain phenotypes. Nature. 2006;442(7102):585‐589. [DOI] [PubMed] [Google Scholar]
- 186. Goldsbury C, Frey P, Olivieri V, Aebi U, Muller SA. Multiple assembly pathways underlie amyloid‐beta fibril polymorphisms. J Mol Biol. 2005;352(2):282‐298. [DOI] [PubMed] [Google Scholar]
- 187. Acquatella‐Tran Van Ba I, Imberdis T, Perrier V. From prion diseases to prion‐like propagation mechanisms of neurodegenerative diseases. Int J Cell Biol. 2013;2013:975832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Telling GC, Parchi P, DeArmond SJ, et al. Evidence for the conformation of the pathologic isoform of the prion protein enciphering and propagating prion diversity. Science. 1996;274(5295):2079‐2082. [DOI] [PubMed] [Google Scholar]
- 189. Petkova AT, Leapman RD, Guo Z, Yau WM, Mattson MP, Tycko R. Self‐propagating, molecular‐level polymorphism in Alzheimer's beta‐amyloid fibrils. Science. 2005;307(5707):262‐265. [DOI] [PubMed] [Google Scholar]
- 190. Poggiolini I, Saverioni D, Parchi P. Prion protein misfolding, strains, and neurotoxicity: an update from studies on mammalian prions. Int J Cell Biol. 2013;2013:910314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Sun JL, Kim S, Crowell J, et al. Novel prion strain as cause of chronic wasting disease in a moose, Finland. Emerg Infect Dis. 2023;29(2):323‐332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Meyer‐Luehmann M, Coomaraswamy J, Bolmont T, et al. Exogenous induction of cerebral beta‐amyloidogenesis is governed by agent and host. Science. 2006;313(5794):1781‐1784. [DOI] [PubMed] [Google Scholar]
- 193. Paravastu AK, Leapman RD, Yau W‐M, Tycko R. Molecular structural basis for polymorphism in Alzheimer’s β‐amyloid fibrils. Proceedings of the National Academy of Sciences. 2008;105(47): 18349–18354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Lo Cascio F, Garcia S, Montalbano M, et al. Modulating disease‐relevant tau oligomeric strains by small molecules. J Biol Chem. 2020;295(44):14807‐14825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Frost B, Diamond MI. Prion‐like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11(3):155‐159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Sanders DW, Kaufman SK, DeVos SL, et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron. 2014;82(6):1271‐1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Liu L, Drouet V, Wu JW, et al. Trans‐synaptic spread of tau pathology in vivo. PloS One. 2012;7(2):e31302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Mudher A, Colin M, Dujardin S, et al. What is the evidence that tau pathology spreads through prion‐like propagation? Acta Neuropathol Commun. 2017;5(1):99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Tycko R. Solid‐state NMR studies of amyloid fibril structure. Annu Rev Phys Chem. 2011;62:279‐299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Schmidt M, Sachse C, Richter W, Xu C, Fandrich M, Grigorieff N. Comparison of Alzheimer Abeta(1‐40) and Abeta(1‐42) amyloid fibrils reveals similar protofilament structures. Proc Natl Acad Sci U S A. 2009;106(47):19813‐19818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Greenwald J, Riek R. Biology of amyloid: structure, function, and regulation. Structure. 2010;18(10):1244‐1260. [DOI] [PubMed] [Google Scholar]
- 202. Kovacs GG. Tauopathies. Handb Clin Neurol. 2017;145:355‐368. [DOI] [PubMed] [Google Scholar]
- 203. Zhang Y, Wu KM, Yang L, Dong Q, Yu JT. Tauopathies: new perspectives and challenges. Mol Neurodegener. 2022;17(1):28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. McFarthing K, Rafaloff G, Baptista M, et al. Parkinson's disease drug therapies in the clinical trial pipeline: 2022 update. J Parkinsons Dis. 2022;12(4):1073‐1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Islam M, Shen F, Regmi D, Du D. Therapeutic strategies for tauopathies and drug repurposing as a potential approach. Biochem Pharmacol. 2022;198:114979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Smit JW, Basile P, Prato MK, et al. Phase 1/1b studies of UCB0599, an oral inhibitor of alpha‐synuclein misfolding, including a randomized study in Parkinson's disease. Mov Disord. 2022;37(10):2045‐2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Knorz AL, Quante A. Alzheimer's disease: efficacy of mono‐ and combination therapy. A systematic review. J Geriatr Psychiatry Neurol. 2022;35(4):475‐486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Cox D, Raeburn C, Sui X, Hatters DM. Protein aggregation in cell biology: an aggregomics perspective of health and disease. Semin Cell Dev Biol. 2020;99:40‐54. [DOI] [PubMed] [Google Scholar]
- 209. Gauthier S, Boxer A, Knopman D, et al. Therapeutic targets for Alzheimer's disease: amyloid vs. non‐amyloid. Where does consensus lie today? An CTAD task force report. J Prev Alzheimers Dis. 2022;9(2):231‐235. [DOI] [PubMed] [Google Scholar]
- 210. Housmans JAJ, Wu G, Schymkowitz J, Rousseau F. A guide to studying protein aggregation. FEBS J. 2023;290(3):554‐583. [DOI] [PubMed] [Google Scholar]
- 211. Imbimbo BP, Ippati S, Watling M, Balducci C. A critical appraisal of tau‐targeting therapies for primary and secondary tauopathies. Alzheimers Dement. 2022;18(5):1008‐1037. [DOI] [PubMed] [Google Scholar]
- 212. Parrocha CT, Nowick JS. Current peptide vaccine and immunotherapy approaches against Alzheimer's disease. Pept Sci. 2022;115(1):e24289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Salloway S, Chalkias S, Barkhof F, et al. Amyloid‐related imaging abnormalities in 2 phase 3 studies evaluating aducanumab in patients with early Alzheimer disease. JAMA Neurol. 2022;79(1):13‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Cingaram PR. A potential therapeutic approach for tauopathies. Mol Ther Nucleic Acids. 2022;30:15‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Patel S, Bansoad AV, Singh R, Khatik GL. BACE1: a key regulator in Alzheimer's disease progression and current development of its inhibitors. Curr Neuropharmacol. 2022;20(6):1174‐1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Sigurdsson EM. Tau immunotherapies for Alzheimer's disease and related tauopathies: progress and potential pitfalls. J Alzheimers Dis. 2018;64(s1):S555‐S565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Advancing Health through Innovation: New Drug Therapy Approvals 2021. Center for Drug Evaluation and Research. US Food & Drug Administration; 2022. Accessed 2022. https://www.fda.gov/media/155227/download [Google Scholar]
- 218. ALZFORUM . Therapeutics‐lecanemab. 2022. https://www.alzforum.org/therapeutics/lecanemab. https://www.alzforum.org/therapeutics/lecanemab
- 219. ALZFORUM . Therapeutics. Aduhelm. https://www.alzforum.org/therapeutics/aduhelm 2022.
- 220. Sevigny J, Chiao P, Bussiere T, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer's disease. Nature. 2016;537(7618):50‐56. [DOI] [PubMed] [Google Scholar]
- 221. Nguyen PH, Ramamoorthy A, Sahoo BR, et al. Amyloid oligomers: a joint experimental/computational perspective on Alzheimer's disease, Parkinson's disease, type II diabetes, and amyotrophic lateral sclerosis. Chem Rev. 2021;121(4):2545‐2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Panza F, Lozupone M, Seripa D, Imbimbo BP. Amyloid‐beta immunotherapy for Alzheimer disease: is it now a long shot? Ann Neurol. 2019;85(3):303‐315. [DOI] [PubMed] [Google Scholar]
- 223. Song C, Shi J, Zhang P, et al. Immunotherapy for Alzheimer's disease: targeting beta‐amyloid and beyond. Transl Neurodegener. 2022;11(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Golde TE. Disease‐modifying therapies for Alzheimer's disease: more questions than answers. Neurotherapeutics. 2022;19(1):209‐227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Wang L, Bharti KR, Pavlov PF, Winblad B. Small molecule therapeutics for tauopathy in Alzheimer's disease: walking on the path of most resistance. Eur J Med Chem. 2021;209:112915. [DOI] [PubMed] [Google Scholar]
- 226. Rayman JB. Focusing on oligomeric tau as a therapeutic target in Alzheimer's disease and other tauopathies. Expert Opin Ther Targets. 2023;27(4‐5):269‐279. [DOI] [PubMed] [Google Scholar]
- 227. Marcatti M, Fracassi A, Montalbano M, et al. Abeta/tau oligomer interplay at human synapses supports shifting therapeutic targets for Alzheimer's disease. Cell Mol Life Sci. 2022;79(4):222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Lo CH. Heterogeneous tau oligomers as molecular targets for Alzheimer's disease and related tauopathies. Biophysica. 2022;2:440‐451. [Google Scholar]
- 229. Huang YR, Liu RT. The toxicity and polymorphism of beta‐amyloid oligomers. Int J Mol Sci. 2020;21:4477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Goedert M, Clavaguera F, Tolnay M. The propagation of prion‐like protein inclusions in neurodegenerative diseases. Trends Neurosci. 2010;33(7):317‐325. [DOI] [PubMed] [Google Scholar]
- 231. Yuan M, Tang X, Han W. Anatomy and formation mechanisms of early amyloid‐beta oligomers with lateral branching: graph network analysis on large‐scale simulations. Chem Sci. 2022;13(9):2649‐2660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Karran E, De Strooper B. The amyloid hypothesis in Alzheimer disease: new insights from new therapeutics. Nat Rev Drug Discov. 2022;21(4):306‐318. [DOI] [PubMed] [Google Scholar]
- 233. Tofaris GK. Initiation and progression of alpha‐synuclein pathology in Parkinson's disease. Cell Mol Life Sci. 2022;79(4):210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. VandeVrede L, Boxer AL, Polydoro M. Targeting tau: clinical trials and novel therapeutic approaches. Neurosci Lett. 2020;731:134919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Fontana IC, Zimmer AR, Rocha AS, et al. Amyloid‐beta oligomers in cellular models of Alzheimer's disease. J Neurochem. 2020;155(4):348‐369. [DOI] [PubMed] [Google Scholar]
- 236. Viola KL, Bicca MA, Bebenek AM, et al. The therapeutic and diagnostic potential of amyloid beta oligomers selective antibodies to treat Alzheimer's disease. Front Neurosci. 2021;15:768646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Lee AK, Yi N, Khaled H, Feller B, Takahashi H. SorCS1 inhibits amyloid‐beta binding to neurexin and rescues amyloid‐beta‐induced synaptic pathology. Life Sci Alliance. 2023;6(4):e202201681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Scannevin RH. Therapeutic strategies for targeting neurodegenerative protein misfolding disorders. Curr Opin Chem Biol. 2018;44:66‐74. [DOI] [PubMed] [Google Scholar]
- 239. Doody RS, Raman R, Farlow M, et al. A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N Engl J Med. 2013;369(4):341‐350. [DOI] [PubMed] [Google Scholar]
- 240. Koriyama Y, Hori A, Ito H, et al. Discovery of atabecestat (JNJ‐54861911): a thiazine‐based beta‐amyloid precursor protein cleaving enzyme 1 inhibitor advanced to the phase 2b/3 EARLY clinical trial. J Med Chem. 2021;64(4):1873‐1888. [DOI] [PubMed] [Google Scholar]
- 241. Neumann U, Ufer M, Jacobson LH, et al. The BACE‐1 inhibitor CNP520 for prevention trials in Alzheimer's disease. EMBO Mol Med. 2018;10(11):e9316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Ugbaja SC, Appiah‐Kubi P, Lawal MM, Gumede NS, Kumalo HM. Unravelling the molecular basis of AM‐6494 high potency at BACE1 in Alzheimer's disease: an integrated dynamic interaction investigation. J Biomol Struct Dyn. 2022;40(12):5253‐5265. [DOI] [PubMed] [Google Scholar]
- 243. Velander P, Wu L, Henderson F, Zhang S, Bevan DR, Xu B. Natural product‐based amyloid inhibitors. Biochem Pharmacol. 2017;139:40‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Santa‐Maria I, Varghese M, Ksiezak‐Reding H, Dzhun A, Wang J, Pasinetti GM. Paired helical filaments from Alzheimer disease brain induce intracellular accumulation of tau protein in aggresomes. J Biol Chem. 2012;287(24):20522‐20533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Miao J, Shi R, Li L, et al. Pathological tau from Alzheimer's brain induces site‐specific hyperphosphorylation and SDS‐ and reducing agent‐resistant aggregation of tau in vivo. Front Aging Neurosci. 2019;11:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Friedhoff P, von Bergen M, Mandelkow EM, Davies P, Mandelkow E. A nucleated assembly mechanism of Alzheimer paired helical filaments. Proc Natl Acad Sci U S A. 1998;95(26):15712‐15717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Kuret J, Chirita CN, Congdon EE, et al. Pathways of tau fibrillization. Biochim Biophys Acta. 2005;1739(2–3):167‐178. [DOI] [PubMed] [Google Scholar]
- 248. Crowther RA. Straight and paired helical filaments in Alzheimer disease have a common structural unit. Proc Natl Acad Sci U S A. 1991;88(6):2288‐2292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Lasagna‐Reeves CA, Castillo‐Carranza DL, Sengupta U, Clos AL, Jackson GR, Kayed R. Tau oligomers impair memory and induce synaptic and mitochondrial dysfunction in wild‐type mice. Mol Neurodegener. 2011;6:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Pasieka A, Panek D, Szalaj N, 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 Nerosci. 2021;12(11):2057‐2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Gadad BS, Britton GB, Rao KS. Targeting oligomers in neurodegenerative disorders: lessons from alpha‐synuclein, tau, and amyloid‐beta peptide. J Alzheimers Dis. 2011;24(Suppl 2):223‐232. [DOI] [PubMed] [Google Scholar]
- 252. Gauthier S, Feldman HH, Schneider LS, et al. Efficacy and safety of tau‐aggregation inhibitor therapy in patients with mild or moderate Alzheimer's disease: a randomised, controlled, double‐blind, parallel‐arm, phase 3 trial. Lancet. 2016;388(10062):2873‐2884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Tang M, Taghibiglou C. The mechanisms of action of curcumin in Alzheimer's disease. J Alzheimers Dis. 2017;58(4):1003‐1016. [DOI] [PubMed] [Google Scholar]
- 254. Liu M, Dexheimer T, Sui D, et al. Hyperphosphorylated tau aggregation and cytotoxicity modulators screen identified prescription drugs linked to Alzheimer's disease and cognitive functions. Sci Rep. 2020;10(1):16551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. VandeVrede L, Ljubenkov PA, Rojas JC, Welch AE, Boxer AL. Four‐repeat tauopathies: current management and future treatments. Neurotherapeutics. 2020;17(4):1563‐1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Chirita C, Necula M, Kuret J. Ligand‐dependent inhibition and reversal of tau filament formation. Biochemistry. 2004;43(10):2879‐2887. [DOI] [PubMed] [Google Scholar]
- 257. Frenkel‐Pinter M, Tal S, Scherzer‐Attali R, et al. Cl‐NQTrp alleviates tauopathy symptoms in a model organism through the inhibition of tau aggregation‐engendered toxicity. Neurodegener Dis. 2017;17(2–3):73‐82. [DOI] [PubMed] [Google Scholar]
- 258. Fuse S, Matsumura K, Fujita Y, Sugimoto H, Takahashi T. Development of dual targeting inhibitors against aggregations of amyloid‐beta and tau protein. Eur J Med Chem. 2014;85:228‐234. [DOI] [PubMed] [Google Scholar]
- 259. Calcul L, Zhang B, Jinwal UK, Dickey CA, Baker BJ. Natural products as a rich source of tau‐targeting drugs for Alzheimer's disease. Future Med Chem. 2012;4(13):1751‐1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. Wilcock GK, Gauthier S, Frisoni GB, et al. Potential of low dose leuco‐methylthioninium bis(hydromethanesulphonate) (LMTM) monotherapy for treatment of mild Alzheimer's disease: cohort analysis as modified primary outcome in a phase III clinical trial. J Alzheimers Dis. 2018;61(1):435‐457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Lee HE, Lim D, Lee JY, Lim SM, Pae AN. Recent tau‐targeted clinical strategies for the treatment of Alzheimer's disease. Future Med Chem. 2019;11(15):1845‐1848. [DOI] [PubMed] [Google Scholar]
- 262. Pasinetti GM, Ksiezak‐Reding H, Santa‐Maria I, Wang J, Ho L. Development of a grape seed polyphenolic extract with anti‐oligomeric activity as a novel treatment in progressive supranuclear palsy and other tauopathies. J Neurochem. 2010;114(6):1557‐1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Price DL, Koike MA, Khan A, et al. The small molecule alpha‐synuclein misfolding inhibitor, NPT200‐11, produces multiple benefits in an animal model of Parkinson's disease. Sci Rep. 2018;8(1):16165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Manzano S, Aguera L, Aguilar M, Olazaran J. A review on tramiprosate (homotaurine) in Alzheimer's disease and other neurocognitive disorders. Front Neurol. 2020;11:614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. van Eersel J, Ke YD, Liu X, et al. Sodium selenate mitigates tau pathology, neurodegeneration, and functional deficits in Alzheimer's disease models. Proc Natl Acad Sci U S A. 2010;107(31):13888‐13893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Cardoso BR, Roberts BR, Malpas CB, et al. Supranutritional sodium selenate supplementation delivers selenium to the central nervous system: results from a randomized controlled pilot trial in Alzheimer's disease. Neurotherapeutics. 2019;16(1):192‐202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. del Ser T, Steinwachs KC, Gertz HJ, et al. Treatment of Alzheimer's disease with the GSK‐3 inhibitor tideglusib: a pilot study. J Alzheimers Dis. 2013;33(1):205‐215. [DOI] [PubMed] [Google Scholar]
- 268. Hoglinger GU, Huppertz HJ, Wagenpfeil S, et al. Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy in a randomized trial. Mov Disord. 2014;29(4):479‐487. [DOI] [PubMed] [Google Scholar]
- 269. Lovestone S, Boada M, Dubois B, et al. A phase II trial of tideglusib in Alzheimer's disease. J Alzheimers Dis. 2015;45(1):75‐88. [DOI] [PubMed] [Google Scholar]
- 270. ClinicalTrials.gov . OLE of phase 2b/3 study ANAVEX2‐73‐AD‐004 (ATTENTION‐AD). ClinicalTrials.gov identifier: NCT04314934. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT04314934?term=ANAVEX2-73&draw=2&rank=2 2020.
- 271. Adelusi TI. Molecular modeling in drug discovery. Inform Med Unlocked. 2022;29:100880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Santos LHS, Ferreira RS, Caffarena ER. Integrating molecular docking and molecular dynamics simulations. Methods Mol Biol. 2019;2053:13‐34. [DOI] [PubMed] [Google Scholar]
- 273. Brogi S, Ramalho TC, Kuca K, Medina‐Franco JL, Valko M. Editorial: in silico methods for drug design and discovery. Front Chem. 2020;8:612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Karplus M, McCammon JA. Molecular dynamics simulations of biomolecules. Nat Struct Biol. 2002;9(9):646‐652. [DOI] [PubMed] [Google Scholar]
- 275. Penney J, Ralvenius WT, Tsai LH. Modeling Alzheimer's disease with iPSC‐derived brain cells. Mol Psychiatry. 2020;25(1):148‐167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Linh NH, Man VH, Li MS, et al. Molecular dynamics simulation of cancer cell membrane perforated by shockwave induced bubble collapse. J Chem Phys. 2022;157(22):225102. [DOI] [PubMed] [Google Scholar]
- 277. Chong SH, Chatterjee P, Ham S. Computer simulations of intrinsically disordered proteins. Annu Rev Phys Chem. 2017;68:117‐134. [DOI] [PubMed] [Google Scholar]
- 278. Nguyen PH, Tuffery P, Derreumaux P. Dynamics of amyloid formation from simplified representation to atomistic simulations. Methods Mol Biol. 2022;2405:95‐113. [DOI] [PubMed] [Google Scholar]
- 279. Nguyen PH, Derreumaux P. An S‐shaped Abeta42 cross‐beta hexamer embedded into a lipid bilayer reveals membrane disruption and permeability. ACS Chem Nerosci. 2023;14(5):936‐946. [DOI] [PubMed] [Google Scholar]
- 280. Gnanaraj C, Sekar M, Fuloria S, et al. In silico molecular docking analysis of Karanjin against Alzheimer's and Parkinson's diseases as a potential natural lead molecule for new drug design, development and therapy. Molecules. 2022;27(9):2834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Borah K, Bora K, Mallik S, Zhao Z. Potential therapeutic agents on Alzheimer's disease through molecular docking and molecular dynamics simulation study of plant‐based compounds. Chem Biodivers. 2023;20(1):e202200684. [DOI] [PubMed] [Google Scholar]
- 282. Wu X, Zheng X, Tang H, et al. A network pharmacology approach to identify the mechanisms and molecular targets of curcumin against Alzheimer disease. Medicine (Baltimore). 2022;101(34):e30194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Biasini E, Faccioli P. Functional, pathogenic, and pharmacological roles of protein folding intermediates. Proteins. 2023;1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Nguyen TH, Nguyen PH, Ngo ST, Derreumaux P. Effect of cholesterol molecules on Abeta1‐42 wild‐type and mutants trimers. Molecules. 2022;27(4):1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Ege D. Action mechanisms of curcumin in Alzheimer's disease and its brain targeted delivery. Materials. 2021;14(12):3332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Ma X, Zhao Y, Yang T, et al. Integration of network pharmacology and molecular docking to explore the molecular mechanism of cordycepin in the treatment of Alzheimer's disease. Front Aging Neurosci. 2022;14:1058780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Nguyen PH, Sterpone F, Derreumaux P. Metastable alpha‐rich and beta‐rich conformations of small Abeta42 peptide oligomers. Proteins. 2023;1‐6. [DOI] [PubMed] [Google Scholar]
- 288. Nguyen PH, Derreumaux P. Insights into the mixture of Abeta24 and Abeta42 peptides from atomistic simulations. J Phys Chem B. 2022;126(50):10689‐10696. [DOI] [PubMed] [Google Scholar]
- 289. Derreumaux P, Nguyen P, Sterpone F. Metastable alpha‐rich and beta‐rich conformations of small Aβ42 peptide oligomers. PROTEINS: Struct Funct Bioinform. 2023;1‐6. [DOI] [PubMed] [Google Scholar]
- 290. Hojati S, Ghahghaei A, Lagzian M. The potential inhibitory effect of beta‐casein on the aggregation and deposition of Abeta(1‐42) fibrils in Alzheimer's disease: insight from in‐vitro and in‐silico studies. J Biomol Struct Dyn. 2018;36(8):2118‐2130. [DOI] [PubMed] [Google Scholar]
- 291. 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):1603‐1611. [DOI] [PubMed] [Google Scholar]
- 292. Murray KA, Hu CJ, Pan H, et al. Small molecules disaggregate alpha‐synuclein and prevent seeding from patient brain‐derived fibrils. Proc Natl Acad Sci U S A. 2023;120(7):e2217835120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Fang M, Zhang Q, Wang X, Su K, Guan P, Hu X. Inhibition mechanisms of (−)‐epigallocatechin‐3‐gallate and genistein on amyloid‐beta 42 peptide of Alzheimer's disease via molecular simulations. ACS Omega. 2022;7(23):19665‐19675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Dufrene YF, Ando T, Garcia R, et al. Imaging modes of atomic force microscopy for application in molecular and cell biology. Nat Nanotechnol. 2017;12(4):295‐307. [DOI] [PubMed] [Google Scholar]
- 295. Pleshakova TO, Bukharina NS, Archakov AI, Ivanov YD. Atomic force microscopy for protein detection and their physicochemical characterization. Int J Mol Sci. 2018;19(4):1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. DiMemmo LM, Cameron Varano A, Haulenbeek J, et al. Real‐time observation of protein aggregates in pharmaceutical formulations using liquid cell electron microscopy. Lab Chip. 2017;17(2):315‐322. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.
