Previous Article in Journal
Adaptation of the Motivational Interviewing Skills Code to Identify Client Language Predicting Reduced Opioid Use Risk and Increased Use of Alternative Pain Care Strategies in Veterans
Previous Article in Special Issue
AgentMol: Multi-Model AI System for Automatic Drug-Target Identification and Molecule Development
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Approach for Targeting Small-Molecule Candidates for Intrinsically Disordered Proteins

Laboratory of Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences Vinca, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia
Methods Protoc. 2025, 8(6), 150; https://doi.org/10.3390/mps8060150
Submission received: 30 October 2025 / Revised: 3 December 2025 / Accepted: 8 December 2025 / Published: 10 December 2025
(This article belongs to the Special Issue Advanced Methods and Technologies in Drug Discovery)

Abstract

Intrinsically disordered proteins (IDPs), such as the Alzheimer’s-associated tau protein, pose challenges for conventional drug discovery. This study applied the Informational Spectrum Method for Small Molecules (ISM-SM), a computational technique utilizing electron–ion interaction potentials (EIIPs), to identify potential tau modulators. Characteristic interaction frequencies derived from known ligands and conserved mammalian tau sequences were used to screen DrugBank and the COCONUT natural product database. The screening identified approved drugs previously reported to indirectly influence tau pathology or Alzheimer’s disease pathways, alongside natural products including Bryostatin-14, which is known to modulate kinases involved in tau phosphorylation. These findings suggest that ISM-SM can serve as an in silico tool to identify candidate small molecules, including repurposed drugs and natural products, with potential relevance to tau function and pathology, complementing other IDP drug discovery strategies.

1. Introduction

Intrinsically disordered proteins (IDPs) play essential roles in a variety of biological processes and have been associated with numerous diseases, including neurodegenerative disorders and viral infections [1,2]. In contrast to structured proteins, which contain stable binding pockets, IDPs exist as dynamic conformational ensembles and represent particularly challenging targets in small-molecule drug discovery. Several small molecules have been identified as IDP ligands, usually by binding to transient interaction sites or as modulators of their dynamic conformational states. Notable examples include epigallocatechin gallate (EGCG) for α-synuclein [3], Phenothiazine for tau [4], and various compounds targeting viral nucleocapsid proteins [5]. IDPs’ inherent flexibility requires specialized experimental and computational methods to identify their ligands.
Conventional structure-based drug development methods are not as successful with IDPs because the binding sites are poorly defined. Similarly, high-throughput virtual screening methods struggle with IDP dynamic and heterogeneous nature. They may call for alternate techniques such as ensemble docking [6,7,8,9,10,11], molecular dynamics (MD) simulations [12,13,14,15,16,17,18,19,20], and machine learning models trained using IDP–ligand interactions [16,21,22,23,24,25]. Experimental methods, including nuclear magnetic resonance (NMR) spectroscopy, surface plasmon resonance (SPR), and fluorescence-based assays, can also help demonstrate weak, transient interactions [26]. Therefore, computational and biophysical methods are crucial for discovering and optimizing small-molecule ligands.
The standard new drug development process typically involves hit identification, lead optimization, preclinical testing, and clinical trials. The cost of bringing a drug to market follows the industry standard, averaging USD 1.39–2.87 billion over 10–15 years, with high attrition rates due to the complexity of validating functional effects and ensuring specificity [27]. Due to the additional complexity of IDP–ligand interactions, this process is more challenging, and the introduction of various in silico methods is required.
The ISM-SM (Informational Spectrum Method for Small Molecules) method offers a distinct approach compared to traditional High-Throughput Screening (HTPS) methods for finding small-molecule candidates for disordered proteins, specifically by analyzing long-range interaction potential rather than relying solely on structure [28]. ISM-SM can put molecular structures into a frequency spectrum, enabling it to identify the compatible interaction frequencies for small molecules and target proteins, predicting biological activity. Our previous ISM-SM studies have successfully identified biologically active ligands for specific protein binding sites [29,30]. ISM-SM has also been utilized to discover binders towards proteins associated with emerging viral threats, such as SARS-CoV-2. An example of this kind is the determination of the interaction frequencies between small molecules that have been shown to be beneficial against viral proteins, predicting drugs that could be repurposed for COVID-19, and significantly accelerating the drug discovery process in response to a pressing public health emergency [31,32,33]. This study explores the application of ISM-SM for identifying potential therapeutic candidates against selected disease targets. We focus on human tau protein candidates, one of the two hallmark proteins of Alzheimer’s disease (AD) [34].

2. Materials and Methods

2.1. Databases

The sequence of Human microtubule-associated protein tau (P10636) was taken from the UniProt database (www.uniprot.org, accessed on 30 October 2025) [35]. Tau proteins from other mammals were also downloaded:
  • O02828 Capra hircus;
  • P10637 Mus musculus;
  • P19332 Rattus norvegicus;
  • P29172 Bos taurus;
  • P57786 Macaca mulatta;
  • Q5S6V2 Pongo pygmaeus;
  • Q5YCV9 Hylobates lar;
  • Q5YCW0 Gorilla gorilla gorilla;
  • Q5YCW1 Pan troglodytes;
  • Q6TS35 Spermophilus citellus;
  • Q9MYX8 Papio hamadryas.
For the screening of drugs for repurposing to select candidates for Microtubule-associated protein tau, 2627 approved small-molecule drugs from DrugBank [36] (http://www.drugbank.ca, accessed on 30 October 2025) were screened. The criteria for candidate selection were five reported tau protein binding drugs: DB00637 Astemizole, DB01248 Docetaxel, DB14914 Flortaucipir F-18, DB00448 Lansoprazole, and DB01229 Paclitaxel.
The COCONUT database [37] is a freely available collection of over six hundred thousand natural products, some of which may also be commercially available (https://coconut.naturalproducts.net/, accessed on 30 October 2025). The COCONUT database was downloaded, and compounds were converted to SMILES notation.

2.2. ISM-SM Method

In this study, we investigate the tau protein and its small-molecule ligands using the Informational Spectrum Method for Small Molecules (ISM-SM), an extension of the Informational Spectrum Method (ISM). This bioinformatics approach enables the analysis of long-range molecular interactions based on the electronic properties encoded in primary structures of biomolecules.
The ISM-SM approach consists of the following key steps:
  • Numerical Encoding of Protein Sequences: The primary amino acid sequence of the protein is converted into a numerical series by assigning each residue its corresponding electron–ion interaction potential (EIIP) value.
  • Numerical Encoding of Small Molecules: The molecular structure of a small molecule, represented in SMILES notation, is translated into a numerical sequence by mapping each atomic group to its EIIP value.
  • Informational Spectrum Calculation: The numerical sequences obtained for the protein and small molecules are transformed into informational spectra (IS) using the discrete Fourier transform (DFT). This process decomposes the sequences into frequencies and amplitudes, revealing periodicities corresponding to structural and functional motifs.
  • Cross-Spectrum (CS) Analysis: The interaction potential between the protein and small molecules is assessed by calculating the cross-spectrum, which identifies shared frequencies in their respective IS profiles. These common frequencies indicate potential sites of interaction or functional correlation.
The electron–ion interaction potential (EIIP) is a physical parameter that reflects the long-range electronic properties of biomolecules, relevant for interactions at distances ranging from 5 to 1000 Å [38,39]. EIIP values are computed using the following equations:
W = 0.25 Z * × s i n ( 1.04 π Z * ) 2 π ,
Z * = 1 N i = 1 m n i Z i
In Equation (2), Z∗ is the average quasi-valence number (AQVN), N is the total number of atoms, ni is the number of atoms of the i-th type, and Zi is the valence of the i-th element. The EIIP values derived from Equation (1) are expressed in Rydbergs (Ry) [40,41].
The discrete Fourier transformation (DFT) used to obtain the IS is defined by
X ( n ) = m = 1 N x m e i 2 π ( m 1 ) / N ,   n = 1 , 2 , , N / 2
Here (3), x(m) is the m-th element of the input numerical sequence of length N, and X(n) is the resulting Fourier coefficient. The frequencies in the IS correspond to biologically relevant periodicities associated with structural or functional motifs in the sequence. To identify shared informational content between molecules, a cross-spectrum (CS) is constructed:
C j = i = 1 N S ( i , j )
In Equation (4), C(j) is the j-th component of the CS, and S(i,j) is the j-th frequency component of the i-th IS. Frequencies with high amplitudes in the CS indicate conserved information relevant to biological function or molecular interaction. The amplitude reflects interaction strength, while the signal-to-noise ratio (S/N) provides a measure of interaction specificity.
Finally, sliding window analysis across the protein sequence allows for the localization of domains contributing to specific intermolecular recognition, enabling the identification of potential binding regions for interacting partners, including proteins and small molecules.

2.3. Drug Score Calculation

Drug Score (dS) values are calculated in DataWarrior [42]. The following descriptors required are calculated (s): druglikeness (d), logP, logS, Molecular Weight (MW), and four types of drug toxicity (t): primary irritation, mutagenic effects, reproductive effects, and tumorigenic effects. The druglikeness in DataWarrior is partially based on topological descriptors, fingerprints of MDL structure keys or other properties as cLogP and molecular weights, including a list of about 5300 distinct substructure fragments with associated druglikeness scores. The druglikeness is calculated using the following Equation (5), summing up score values of those fragments that are present in the molecule under investigation:
d = v i n
The drug score (dS) is calculated according to the following formulas (6) and (7):
d S = ( 1 2 + 1 2 s i ) · t i
s i = 1 1 + e a p + b
where p corresponds to logP, logS, MW and d, and parameters a and b correspond to values {1, −5}, {1, 5}, {0.012, 6}, {1, 0}, respectively. The ti values are 1.0, 0.8 and 0.6 for no risk, low risk, and high risk, respectively. A detailed reference on the calculation of molecular properties can be found in the DataWarrior manual at https://openmolecules.org/properties/properties.html (accessed on 30 October 2025).
The Total Score had to incorporate long-range interaction properties, measured according to the ISM-SM signal-to-noise ratio (S/N) and Drug Score. As both parameters favor the higher values, the Total Score is therefore calculated as their product (8), i.e.,
T o t a l   S c o r e = d S × S / N
The full methodology workflow is presented in Scheme 1.

2.4. Pharmacokinetics Predictions

The pharmacokinetic properties of the compounds were calculated using the pkCSM [43] server (https://biosig.lab.uq.edu.au/pkcsm/prediction, accessed on 25 November 2025), with a total number of variables of 36. The detailed interpretation of calculated ADMET properties is available at https://biosig.lab.uq.edu.au/pkcsm/theory (accessed on 25 November 2025). In the case of non-numerical prediction results Yes/No, for further calculations, they were converted to 1 and 0, respectively. The resulting data were subjected to k-means clustering.

2.5. Continuous Wavelet Transform (CWT)

In this study, we use CWT to identify hotspot amino acid residues in a protein sequence at a specific ISM frequency. The detailed procedure is described in our previous work [44].

3. Results and Discussion

3.1. DrugBank Candidates

Five reported drugs from the Uniprot database targeting Microtubule-associated protein tau protein (P10636) were extracted with their structures in SMILES format (Figure 1). The structures were converted into the explicit hydrogen format, and their CS with the tau protein were calculated, along with the corresponding interaction domains (Table 1). Five frequencies, F(0.080), F(0.167), F(0.194), F(0.342) and F(0.435), were identified (Figure 2). The additional frequency from CS, including all five drugs and tau protein, was found at F(0.333) (Figure 3). Interestingly, the same frequency was obtained from the CS of all mammal tau proteins (Figure 4). This suggests an evolutionarily conserved region in the tau protein.
Regarding the binding domains of tau protein, Astemizole, Flortaucipir F-18 and Lansoprazole bind to R3-R4 regions [45,46,47], while Docetaxel and Paclitaxel do not bind directly to tau, but β-tubulin. The calculated domains corresponding to CS frequencies agree with the literature, even indirectly, due to the partial spanning of the Microtubule-Binding (MTBD) region (244–368) [48] (Figure 5). The region corresponding to F (0.333) corresponds to the residues 494–750. This C-terminal domain is not directly responsible for microtubule binding, but rather it may modulate or influence this interaction indirectly under certain conditions, such as phosphorylation or aggregation [49].
The 2627 approved DrugBank candidates’ structures were subjected to the exact format conversion as literature compounds and were further CS-scanned at all six frequencies. Of the candidates obtained (Table S1), 19 were already reported to indirectly affect the tau protein or AD progression (Table 2, Figure 6).
Cinacalcet was reported to play a significant role in AD. As a calcimimetic agent used for hyperparathyroidism, it modulates calcium-sensing receptors, which may influence amyloid-beta (Aβ) pathology and neuronal calcium dysregulation, both implicated in AD. It may indirectly influence tau phosphorylation by regulating calcium signaling, but direct evidence is lacking [50]. Bezafibrate is a PPAR-alpha agonist used for lipid disorders. It has been shown that bezafibrate treatment could attenuate the severity of tau pathology in the streptozotocin-intracerebroventricular-induced sporadic AD rat model [51]. Temsirolimus [52,53] and Everolimus [54,55] were reported to demonstrate neuroprotective effects in AD models by reducing tau hyperphosphorylation and promoting autophagic clearance of amyloid-β (Aβ) and tau aggregates, improving cognitive function. Desmopressin [56], a neurohypophyseal hormone analog, has been suggested to modulate amyloid aggregation, though its direct role in tau pathology remains less explored. Prednicarbate [57] (a topical corticosteroid) was identified in a drug screening study as one of the prescription drugs that may influence hyperphosphorylated tau aggregation and cytotoxicity. Roflumilast [58] ameliorates cognitive deficits in AD mice by reducing Aβ and tau pathology, potentially via nitric oxide signaling and upregulating Aβ transporters such as ABCB1. Lipoic acid [59,60,61,62,63,64,65,66,67,68] shows potent antioxidant and anti-inflammatory properties, mitigating tau hyperphosphorylation, oxidative stress, and behavioral deficits in tauopathy models, while enhancing mitochondrial function. Testosterone Propionate is a synthetic androgen, and androgens are found to regulate tau phosphorylation [69]. Cabazitaxel is a chemotherapy agent. While not directly linked to AD, its ability to stabilize microtubules has prompted interest in its potential to address tau pathology, a hallmark of AD [70]. Probucol is a lipid-lowering drug with antioxidant properties. It may reduce oxidative stress and amyloid deposition, which are implicated in AD [71]. Estradiol valerate and Dienogest are hormonal agents. Estrogen has been studied for its neuroprotective effects, preventing neural tau hyperphosphorylation, particularly in postmenopausal women, who are at higher risk for AD [72]. Perphenazine is an antipsychotic, found, among some others, to lower the levels of insoluble Tau [73]. Degarelix is a GnRH antagonist. Hormonal modulation has been explored in AD, particularly regarding sex hormones and their impact on cognitive function [74]. Isosorbide Dinitrate is a vasodilator. Improving cerebral blood flow may have neuroprotective effects in AD [75]. Ranolazine is an antianginal drug. It modulates cellular metabolism and has been explored for its potential to enhance neuronal energy deficits in AD [76]. Methocarbamol [77], a carbonic anhydrase inhibitor, has been shown to reduce tau toxicity by promoting its clearance. Studies in tauopathy models, including zebrafish and transgenic mice, have demonstrated that methocarbamol can rescue neuronal degeneration, improve cognitive function, and reduce phosphorylated tau levels. Carvedilol is a beta-blocker with antioxidant properties. It may reduce oxidative stress and inflammation, both implicated in AD [78]. Reserpine is an antihypertensive with neuroprotective potential. Reserpine [79,80], in particular, has been studied for its ability to modulate neurotransmitter systems involved in AD. The complete list of the DrugBank candidates is given in the Table S2.
Although not directly involved in interactions with tau protein, the identified drugs affect processes in AD via other targets in the tau signaling pathway, such as phosphorylation. This may be possible due to the PPI interactions in the signaling pathways, occurring at the standard ISM frequency. This suggests the method might capture broader signaling relationships, potentially reflecting in vivo activity, although the mechanism for this requires further investigation and validation.

3.2. COCONUT Database Candidates

Compounds from the COCONUT database were also screened on all frequencies, as were the DrugBank compounds. However, in the case of small organic molecules, unlike the proteins, mere calculation of A and S/N values cannot be considered the final step. Further insight into the candidate’s structure and properties is required. Therefore, the dS values of the candidates were also calculated. Those values were integrated into the final Total Score descriptor as their product, and the candidates were finally sorted accordingly. The top compounds at all frequencies are presented in Table 3.
We identified a highly ranking compound, bryostatin-14, at F(0.167) (Figure 7). Bryostatins are macrocyclic lactones from marine bryozoans. They are increasingly being considered for therapeutic development in AD because of their ability to modulate protein kinase C (PKC) activity and ultimately mitigate pathological features of AD, such as tau hyperphosphorylation and amyloid-β (Aβ) aggregation. Several preclinical studies have demonstrated that bryostatin-1 enhanced synaptic plasticity and cognitive behavior through the activation of PKCε, which inactivated glycogen synthase kinase-3β (GSK-3β), a critical kinase driving tau hyperphosphorylation [81,82]. The inhibition of GSK-3β reduces the pathological aggregation of tau, which improves neurons’ survival in transgenic tauopathy models [81]. Although their binding to tau protein is not well characterized, their modulation of tau phosphorylation through PKC/GSK-3β signaling makes them an unusual therapeutic approach to targeting tauopathies such as AD. Interestingly, bryostatin-14 is a hit at F(0.167), the most populated F among DrugBank compounds (Figure 6).

3.3. Pharmacokinetic Properties of the Selected Compounds

To further assess the druglikeness of the selected compounds, pharmacokinetics calculations were carried out for all three sets of compounds: Tau-interacting drugs (Table 1), DrugBank-identified compounds (Table 2) and COCONUT database candidates (Table 3). ADMET properties were calculated using the pkCSM server [43] (https://biosig.lab.uq.edu.au/pkcsm/prediction, accessed on 25 November 2025), with a total number of variables 36. The resulting data were subjected to k-means clustering.

k-Means Clustering

The number of classes was set to 5 and 10 repetitions. The within-class variance was 4.77%, with a between-class variance of 95.23%. Based on the class centroids (Table S2), the five clusters can be interpreted as follows, according to their distinguishing features:
Cluster 1 consists of the smallest molecules (mean molar weight ≈ 410) with the highest lipophilicity (logP ≈ 4.4), BBB permeability (≈−0.28) and CNS permeability (−2.4) among all clusters. These compounds also displayed favorable Caco-2 permeability (0.862), high intestinal absorption (mean value 88%), and high total clearance (logCLtot = 0.571 mL/min/kg). No significant toxicity is predicted, except for hERG II inhibition (around 40% are recognized). The cluster contains the following DrugBank drugs: Astemizole, a direct Tau-interacting compound, Flortaucipir F-18, Lansoprazole, DB01012 (Cinacalcet), DB01393 (Bezafibrate), DB01130 (Prednicarbate), DB01656 (Roflumilast), DB00166 (Lipoic acid), DB01420 (Testosterone Propionate), DB0159 (Probucol), DB08866 (Estradiol valerate), DB00850 (Perphenazine), DB00883 (Isosorbide Dinitrate), DB00243 (Ranolazine), DB00423 (Methocarbamol) and DB01136 (Carvedilol). Fifteen COCONUT candidates belong to this cluster. Regarding best candidates, CNP0327834.1 (#5 rank at F(0.167), Table 3) is the nearest compound to both Astemizol and Lansoprazole. On the other hand, CNP0151916.0 (ranked #8 at F(0.435)) is the nearest one to the DrugBank candidate group in the cluster. The central compound of the cluster is DB01136 (Carvedilol). Members of this group represent the most conventional “drug-like” space.
Cluster 2 contains molecules with intermediate molecular weight (mean MW ≈ 750), moderate lipophilicity (logP ≈ 3.0), and lower BBB permeability characteristics (≈−2.18). These compounds exhibit good intestinal absorption (≈62%) and lower propensity for CNS entry (≈−3.428), although not to the extent of Cluster 1. This cluster includes several clinically approved agents, Docetaxel and Paclitaxel, along with other DrugBank compounds: DB00206 (Reserpine) and DB06772 (Cabazitaxel). Thirteen COCONUT candidates are found in this cluster. CNP0429159.1 (#10 ranked at F(0.435)) is the nearest to Docetaxel, CNP0423521.1 to Pacitaxel, and the central compound of the cluster is CNP0337940.1 (#10 rank at F(0.435)). While still within the expanded drug-like space, diminished BBB and CNS permeability implies primarily peripheral exposure.
Cluster 3 is characterized by higher MW (≈1000) and moderate lipophilicity, similar to cluster 2 (logP ≈ 2.77). Their physicochemical properties indicate lower intestinal absorption, negative Caco2 permeability, and significantly lower BBB (−3.313) and CNS permeability (−4.505) in comparison to Clusters 1 and 2. The compounds also show a higher number of hERG II inhibitors (60%) and higher hepatotoxicity. Total clearance is also significantly lower. This cluster contains two DrugBank compounds, DB06287 (Temsirolimus) and DB01590 (Everolimus). The central compound of the cluster is CNP0180487.0 (#6 ranked at (0.341).
Cluster 4 molecules possess the heaviest mass (MW 1770), extremely low lipophilicity, and extremely low Caco-2 permeability and absorption. Toxicity is on the rise in this cluster. Despite some structural similarity to approved therapeutics, their ADMET profile indicates high development risk. Only one DrugBank compound, DB06699 (Degarelix), belongs to this cluster.
Cluster 5 is the last one, and molecules there show similar properties to those in Cluster 4, with some minor advantages, such as better Caco-2 and BBB permeability, and total clearance, but these values are still far from Clusters 1 and 2. No approved compounds belong to this cluster.
Overall, the k-means analysis separated the dataset into five distinct ADME-driven chemotypes, ranging from CNS-penetrant lipophilic drugs (Cluster 1) to highly complex, permeability-limited compounds (Clusters 4 and 5), with intermediate clusters reflecting balanced or partially drug-like physicochemical profiles. Regarding the COCONUT database candidates, from the table of centroid distances for each cluster, the following best candidates per cluster are presented in Table S2.
An interesting correlation is found between S/N and Drug Score, where R2 = 0.589 (Figure 8). This is expected, as S/N depends on the amplitude, which, again, depends on the size of the molecule. Larger candidate molecules exhibit a stronger affinity towards proteins. However, the larger the molecule, the less druggable it is, as is also shown in Figure 8. Regardless of F, S/N correlates with dS. We emphasize that dS is the Drug Score, derived from only four properties of a molecule: logP, logS, druggability and molecular weight. Candidates belonging to Cluster 1 (red), the one with best ADMET properties for a small drug molecule, are placed in the area with medium S/N values (20–40) and higher dS values (0.3–0.45). This suggests that, due to negative correlation between S/N and dS, a good candidate should compromise both properties in the Total Score.
By combining S/N values with distance to centroid in each cluster, we can construct another score—Total Score 2. Since the new score should favor higher S/N and lower distance, it should be a ratio of these two values. Therefore, we can rank the compounds per cluster (Table 4). According to the ranking, the candidate with best Total Score 2 is, as already mentioned, CNP0151916.0, a compound with an aporphine structure. It has been reported that aporphines show potential in treating AD, primarily due to their ability to act as cholinesterase inhibitors, similar to some current AD medications. Some aporphines also show promise by reducing oxidative stress and inhibiting β-amyloid formation, which are other key processes in AD [83,84,85,86,87].

3.4. Comparison to the Martini-IDP Forcefield

In the recent study conducted by Wang et al. [19], the Martini forcefield was updated with the parameters for small molecules. The authors carried out 15 molecular dynamics simulations for the validation. Their study examined the contact frequency between the small molecule–ligand and the IDP. The systems studied were Alpha synuclein, p53, and Androgen receptor, as well as their corresponding ligands (see [19]). The corresponding domains with the highest contribution to protein–ligand ISM-SM CS frequencies (Figure 9), and their comparison to the Martini-IDP forcefield, are presented in Table 5.
The results demonstrate that ISM-SM complements traditional forcefield-based approaches, offering alternative insights into IDP-ligand binding regions. The observed overlaps support ISM-SM’s validity, while the method’s broader residue selection suggests it could be particularly valuable for capturing dynamic, transient interactions—hallmarks of intrinsically disordered proteins.

3.5. Comparison to Ensemble Docking Results

A similar study on the ensemble docking of three compounds to alpha-synuclein has been reported recently [7]. Fasudil, Ligand 47 and Ligand 23 were docked to a-synuclein conformations obtained from the MD simulations. According to the results, the amino acid residues with the highest probability for interaction with the ligands are in the domain 121–139, with the top values at Y133 and E137. The ISM-SM analysis confirms this. In Table 6 are presented the top three peaks for each protein–ligand interaction, and corresponding protein domains from the slide window analysis. Finally, Figure 10 presents the common frequency from the CS of the protein and all three ligands, as well as their CS.
To narrow the interaction domain and identify interaction hotspots, a continuous wavelet transform of the alpha-synuclein sequence was performed, similar to our previous study [44]. The heatmaps of direct wavelet coefficient values are presented in Figure 11 and Figure 12, in five shades. Notably, the “hotspot” amino acid residues are in the range of 100–122 (Figure 12). This is in good accordance with the ensemble docking results. Therefore, the ISM-SM method can successfully predict and interpret interaction domains of protein–ligand pairs, based on their sequences, as effectively as structure-based methods.

4. Conclusions

In this study, we examined the applicability and reliability of the ISM-SM method in identifying small-molecule candidates relevant to IDPs, taking human tau protein as a test case. Unlike many other methods, ISMSM does not assume predefined 3D structures but relies on EIIP-based spectral compatibility in order to detect long-range interaction features. Although conceptually different from structure-based approaches, several lines of evidence are presented showing that the method yields biologically meaningful results.
First, ISM-SM recovered a set of approved drugs already reported to modulate tau pathology or Alzheimer’s-related signaling pathways. Although many of these compounds act indirectly, their consistent enrichment at characteristic ISM frequencies supports the method’s ability to capture functionally relevant interaction signatures. Second, the identified natural-product candidates, such as bryostatin-14 and aporphines, show coherence with known tau-related pathways, further supporting the biological relevance of ISM frequencies. Third, comparison with Martini-IDP simulations and ensemble docking revealed overlaps in residues contributing to interaction hotspots. These findings suggest that ISM-SM captures elements of the dynamic recognition patterns typical for IDPs, complementing force-field- and structure-based methods.
At the same time, it is important to recognize the limitations of the method. First, ISM-SM predicts compatibility in the informational spectrum rather than physical binding, and thus cannot directly confirm binding affinity, binding pose, or conformational stabilization. Its use is therefore most reliable as an early-stage screening and hypothesis-generation tool, with experimental validation or detailed structural simulations remaining essential for mechanism-level interpretation. Furthermore, while the ability of ISM-SM to identify compounds acting indirectly through signaling pathways may be an advantage, careful interpretation and further theoretical clarification are required.
Overall, the current results position ISM-SM as a robust and computationally efficient complementary method to explore chemical space around IDP targets. Its strengths include screening large databases at high speeds and the identification of conserved interaction frequencies. Prioritization of candidates based on their favorable pharmacokinetic properties can be easily implemented. Appropriately, together with structural, biophysical, and machine learning methods, ISM-SM can expand the toolbox currently available for rational ligand discovery against dynamic and structurally heterogeneous protein targets.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/mps8060150/s1. Table S1: contains the lists of DrugBank and COCONUT database candidates at all ISM frequencies, along with the amino acid and atomic group EIIP parameters used in the calculations. Table S2: contains the ADMET properties of all compounds, k-means clustering and their ranking by Total Score 2.

Funding

This research was funded by the Ministry of Science, Technological Development and Innovation of the Republic of Serbia, grant number 451-03-136/2025-03/200017.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This article and the Supplementary Material include the original contributions presented in this study. Further inquiries can be directed to the corresponding author.

Acknowledgments

During the preparation of this manuscript/study, the author(s) used ChatGPT–5.1 and Grammarly for the purposes of improving the clarity and language of the manuscript. The authors have reviewed and edited the output and take full responsibility for the content of this publication. The authors are grateful to Vladimir Perović for the software, used in this study to carry out the bioinformatics calculations.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
IDPIntrinsically disordered protein
EGCGEpigallocatechin gallate
MDMolecular Dynamics
NMRNuclear Magnetic Resonance
SPRSurface Plasmon Resonance
ISM-SMInformational Spectrum Method for Small Molecules
HTPSHigh-Throughput Screening
ADAlzheimer’s disease
EIIPElectron Ion Interaction Potential
DFTDiscrete Fourier Transformation
ISInformational Spectra
CSCross Spectrum
MWMolecular Weight
dSDrug Score
S/NSignal-to-Noise ratio
MTBDMicrotubule-Binding
Amyloid-Beta
PKCProtein Kinase C
GSKGlycogen Synthase Kinase
CWTContinuous Wavelet Transform

References

  1. Uversky, V.N. Intrinsically Disordered Proteins and Their (Disordered) Proteomes in Neurodegenerative Disorders. Front. Aging Neurosci. 2015, 7, 18. [Google Scholar] [CrossRef]
  2. Wright, P.E.; Dyson, H.J. Intrinsically Disordered Proteins in Cellular Signalling and Regulation. Nat. Rev. Mol. Cell Biol. 2015, 16, 18–29. [Google Scholar] [CrossRef]
  3. Bieschke, J.; Russ, J.; Friedrich, R.P.; Ehrnhoefer, D.E.; Wobst, H.; Neugebauer, K.; Wanker, E.E. EGCG Remodels Mature α-Synuclein and Amyloid-β Fibrils and Reduces Cellular Toxicity. Proc. Natl. Acad. Sci. USA 2010, 107, 7710–7715. [Google Scholar] [CrossRef]
  4. Akoury, E.; Pickhardt, M.; Gajda, M.; Biernat, J.; Mandelkow, E.; Zweckstetter, M. Mechanistic Basis of Phenothiazine-Driven Inhibition of Tau Aggregation. Angew. Chem. Int. Ed. Engl. 2013, 52, 3511–3515. [Google Scholar] [CrossRef]
  5. Kumari, S.; Mistry, H.; Bihani, S.C.; Mukherjee, S.P.; Gupta, G.D. Unveiling Potential Inhibitors Targeting the Nucleocapsid Protein of SARS-CoV-2: Structural Insights into Their Binding Sites. Int. J. Biol. Macromol. 2024, 273, 133167. [Google Scholar] [CrossRef] [PubMed]
  6. Verburgt, J.; Zhang, Z.; Kihara, D. Multi-Level Analysis of Intrinsically Disordered Protein Docking Methods. Methods 2022, 204, 55–63. [Google Scholar] [CrossRef] [PubMed]
  7. Dhar, A.; Sisk, T.R.; Robustelli, P. Ensemble Docking for Intrinsically Disordered Proteins 2025. J. Chem. Inf. Model. 2025, 65, 6847–6860. [Google Scholar] [CrossRef]
  8. Karatzas, P.; Brotzakis, Z.F.; Sarimveis, H. Small Molecules Targeting the Structural Dynamics of AR-V7 Partially Disordered Proteins Using Deep Ensemble Docking. J. Chem. Theory Comput. 2025, 21, 4898–4909. [Google Scholar] [CrossRef] [PubMed]
  9. Patel, K.N.; Chavda, D.; Manna, M. Molecular Docking of Intrinsically Disordered Proteins: Challenges and Strategies. In Pro-tein-Protein Docking; Methods in Molecular Biology; Kaczor, A.A., Ed.; Springer US: New York, NY, USA, 2024; Volume 2780, pp. 165–201. ISBN 978-1-0716-3984-9. [Google Scholar]
  10. Ruan, H.; Sun, Q.; Zhang, W.; Liu, Y.; Lai, L. Targeting Intrinsically Disordered Proteins at the Edge of Chaos. Drug Discov. Today 2019, 24, 217–227. [Google Scholar] [CrossRef]
  11. Wang, X.; Chong, B.; Sun, Z.; Ruan, H.; Yang, Y.; Song, P.; Liu, Z. More Is Simpler: Decomposition of LIGAND-BINDING Affinity for Proteins Being Disordered. Protein Sci. 2022, 31, e4375. [Google Scholar] [CrossRef]
  12. Apicella, A.; Marascio, M.; Colangelo, V.; Soncini, M.; Gautieri, A.; Plummer, C.J.G. Molecular Dynamics Simulations of the Intrinsically Disordered Protein Amelogenin. J. Biomol. Struct. Dyn. 2017, 35, 1813–1823. [Google Scholar] [CrossRef]
  13. Dobrev, V.S.; Fred, L.M.; Gerhart, K.P.; Metallo, S.J. Characterization of the Binding of Small Molecules to Intrinsically Disordered Proteins. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 2018; Volume 611, pp. 677–702. ISBN 978-0-12-815649-0. [Google Scholar]
  14. Heller, G.T.; Shukla, V.K.; Figueiredo, A.M.; Hansen, D.F. Picosecond Dynamics of a Small Molecule in Its Bound State with an Intrinsically Disordered Protein. J. Am. Chem. Soc. 2024, 146, 2319–2324. [Google Scholar] [CrossRef]
  15. Heller, G.T.; Sormanni, P.; Vendruscolo, M. Targeting Disordered Proteins with Small Molecules Using Entropy. Trends Biochem. Sci. 2015, 40, 491–496. [Google Scholar] [CrossRef]
  16. Herrera-Nieto, P.; Pérez, A.; De Fabritiis, G. Small Molecule Modulation of Intrinsically Disordered Proteins Using Molecular Dynamics Simulations. J. Chem. Inf. Model. 2020, 60, 5003–5010. [Google Scholar] [CrossRef]
  17. Ruan, H.; Yu, C.; Niu, X.; Zhang, W.; Liu, H.; Chen, L.; Xiong, R.; Sun, Q.; Jin, C.; Liu, Y.; et al. Computational Strategy for In-trinsically Disordered Protein Ligand Design Leads to the Discovery of P53 Transactivation Domain I Binding Compounds That Activate the P53 Pathway. Chem. Sci. 2021, 12, 3004–3016. [Google Scholar] [CrossRef] [PubMed]
  18. Vagrys, D.; Davidson, J.; Chen, I.; Hubbard, R.E.; Davis, B. Exploring IDP–Ligand Interactions: Tau K18 as a Test Case. Int. J. Mol. Sci. 2020, 21, 5257. [Google Scholar] [CrossRef] [PubMed]
  19. Wang, L.; Brasnett, C.; Borges-Araújo, L.; Souza, P.C.T.; Marrink, S.J. Martini3-IDP: Improved Martini 3 Force Field for Disor-dered Proteins. Nat. Commun. 2025, 16, 2874. [Google Scholar] [CrossRef]
  20. Papadourakis, M.; Cournia, Z.; Mey, A.S.J.S.; Michel, J. Comparison of Methodologies for Absolute Binding Free Energy Cal-culations of Ligands to Intrinsically Disordered Proteins. J. Chem. Theory Comput. 2024, 20, 9699–9707. [Google Scholar] [CrossRef] [PubMed]
  21. Michel, J.; Cuchillo, R. The Impact of Small Molecule Binding on the Energy Landscape of the Intrinsically Disordered Protein C-Myc. PLoS ONE 2012, 7, e41070. [Google Scholar] [CrossRef]
  22. Wang, H.; Xiong, R.; Lai, L. Rational Drug Design Targeting Intrinsically Disordered Proteins. WIREs Comput. Mol. Sci. 2023, 13, e1685. [Google Scholar] [CrossRef]
  23. Appadurai, R.; Koneru, J.K.; Bonomi, M.; Robustelli, P.; Srivastava, A. Clustering Heterogeneous Conformational Ensembles of Intrinsically Disordered Proteins with T-Distributed Stochastic Neighbor Embedding. J. Chem. Theory Comput. 2023, 19, 4711–4727. [Google Scholar] [CrossRef]
  24. Kattuparambil, A.A.; Chaurasia, D.K.; Shekhar, S.; Srinivasan, A.; Mondal, S.; Aduri, R.; Jayaram, B. Exploring Chemical Space for “Druglike” Small Molecules in the Age of AI. Front. Mol. Biosci. 2025, 12, 1553667. [Google Scholar] [CrossRef] [PubMed]
  25. Lama, D.; Vosselman, T.; Sahin, C.; Liaño-Pons, J.; Cerrato, C.P.; Nilsson, L.; Teilum, K.; Lane, D.P.; Landreh, M.; Arsenian Henriksson, M. A Druggable Conformational Switch in the C-MYC Transactivation Domain. Nat. Commun. 2024, 15, 1865. [Google Scholar] [CrossRef]
  26. Fawzi, N.L.; Ying, J.; Torchia, D.A.; Clore, G.M. Kinetics of Amyloid β Monomer-to-Oligomer Exchange by NMR Relaxation. J. Am. Chem. Soc. 2010, 132, 9948–9951. [Google Scholar] [CrossRef]
  27. DiMasi, J.A.; Grabowski, H.G.; Hansen, R.W. Innovation in the Pharmaceutical Industry: New Estimates of R&D Costs. J. Health Econ. 2016, 47, 20–33. [Google Scholar] [CrossRef] [PubMed]
  28. Sencanski, M.; Sumonja, N.; Perovic, V.; Glisic, S.; Veljkovic, N.; Veljkovic, V. Application of Information Spectrum Method on Small Molecules and Target Recognition. arXiv 2019, arXiv:1907.02713v3. [Google Scholar]
  29. Sencanski, M.; Perovic, V.; Pajovic, S.B.; Adzic, M.; Paessler, S.; Glisic, S. Drug Repurposing for Candidate SARS-CoV-2 Main Protease Inhibitors by a Novel in Silico Method. Molecules 2020, 25, 3830. [Google Scholar] [CrossRef]
  30. Sencanski, M.; Perovic, V.; Milicevic, J.; Todorovic, T.; Prodanovic, R.; Veljkovic, V.; Paessler, S.; Glisic, S. Identification of SARS-CoV-2 Papain-like Protease (PLpro) Inhibitors Using Combined Computational Approach. Chem. Open 2022, 11, 248. [Google Scholar] [CrossRef] [PubMed]
  31. Đukić, I.; Kaličanin, N.; Sencanski, M.; Pajovic, S.B.; Milicevic, J.; Prljic, J.; Paessler, S.; Prodanović, R.; Glisic, S. Inhibition of SARS-CoV-2 Mpro with Vitamin C, L-Arginine and a Vitamin C/L-Arginine Combination. Front. Biosci. 2023, 28, 8. [Google Scholar] [CrossRef]
  32. Protić, S.; Kaličanin, N.; Sencanski, M.; Prodanović, O.; Milicevic, J.; Perovic, V.; Paessler, S.; Prodanović, R.; Glisic, S. In Silico and In Vitro Inhibition of SARS-CoV-2 PLpro with Gramicidin D. Int. J. Mol. Sci. 2023, 24, 1955. [Google Scholar] [CrossRef]
  33. Protić, S.; Crnoglavac Popović, M.; Kaličanin, N.; Prodanović, O.; Senćanski, M.; Milićević, J.; Stevanović, K.; Perović, V.; Paessler, S.; Prodanović, R.; et al. SARS-CoV-2 PLpro Inhibition: Evaluating in Silico Repurposed Fidaxomicin’s Antiviral Activity Through In Vitro Assessment. Chem. Open 2024, 13, e202400091. [Google Scholar] [CrossRef]
  34. Thal, D.R.; Tomé, S.O. The Central Role of Tau in Alzheimer’s Disease: From Neurofibrillary Tangle Maturation to the Induction of Cell Death. Brain Res. Bull. 2022, 190, 204–217. [Google Scholar] [CrossRef]
  35. The UniProt Consortium; Bateman, A.; Martin, M.-J.; Orchard, S.; Magrane, M.; Adesina, A.; Ahmad, S.; Bowler-Barnett, E.H.; Bye-A-Jee, H.; Carpentier, D.; et al. UniProt: The Universal Protein Knowledgebase in 2025. Nucleic Acids Res. 2025, 53, D609–D617. [Google Scholar] [CrossRef]
  36. Wishart, D.S.; Feunang, Y.D.; Guo, A.C.; Lo, E.J.; Marcu, A.; Grant, J.R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; et al. DrugBank 5.0: A Major Update to the DrugBank Database for 2018. Nucleic Acids Res. 2018, 46, D1074–D1082. [Google Scholar] [CrossRef]
  37. Chandrasekhar, V.; Rajan, K.; Kanakam, S.R.S.; Sharma, N.; Weißenborn, V.; Schaub, J.; Steinbeck, C. COCONUT 2.0: A Comprehensive Overhaul and Curation of the Collection of Open Natural Products Database. Nucleic Acids Res. 2025, 53, D634–D643. [Google Scholar] [CrossRef]
  38. Veljković, V.; Cosić, I. A Novel Method of Protein Analysis for Prediction of Biological Function: Application to Tumor Toxins. Cancer Biochem. Biophys. 1987, 9, 139–148. [Google Scholar]
  39. Lechelon, M.; Meriguet, Y.; Gori, M.; Ruffenach, S.; Nardecchia, I.; Floriani, E.; Coquillat, D.; Teppe, F.; Mailfert, S.; Marguet, D.; et al. Experimental Evidence for Long-Distance Electrodynamic Intermolecular Forces. Sci. Adv. 2022, 8, eabl5855. [Google Scholar] [CrossRef]
  40. Veljković, V. The Dependence of the Fermi Energy on the Atomic Number. Phys. Lett. A 1973, 45, 41–42. [Google Scholar] [CrossRef]
  41. Veljković, V.; Slavić, I. Simple General-Model Pseudopotential. Phys. Rev. Lett. 1972, 29, 105–107. [Google Scholar] [CrossRef]
  42. Sander, T.; Freyss, J.; Von Korff, M.; Rufener, C. DataWarrior: An Open-Source Program for Chemistry Aware Data Visuali-zation and Analysis. J. Chem. Inf. Model. 2015, 55, 460–473. [Google Scholar] [CrossRef] [PubMed]
  43. Pires, D.E.V.; Blundell, T.L.; Ascher, D.B. pkCSM: Predicting Small-Molecule Pharmacokinetic and Toxicity Properties Using Graph-Based Signatures. J. Med. Chem. 2015, 58, 4066–4072. [Google Scholar] [CrossRef]
  44. Isaković, S.; Senćanski, M.; Perović, V.; Stevanović, K.; Prodić, I. Bioinformatic Selection of Mannose-Specific Lectins from Allium Genus as SARS-CoV-2 Inhibitors Analysing Protein–Protein Interaction. Life 2025, 15, 162. [Google Scholar] [CrossRef]
  45. Burnham, S.C.; Iaccarino, L.; Pontecorvo, M.J.; Fleisher, A.S.; Lu, M.; Collins, E.C.; Devous, M.D. A Review of the Flortaucipir Literature for Positron Emission Tomography Imaging of Tau Neurofibrillary Tangles. Brain Commun. 2023, 6, fcad305. [Google Scholar] [CrossRef] [PubMed]
  46. Rojo, L.E.; Alzate-Morales, J.; Saavedra, I.N.; Davies, P.; Maccioni, R.B. Selective Interaction of Lansoprazole and Astemizole with Tau Polymers: Potential New Clinical Use in Diagnosis of Alzheimer’s Disease. JAD 2010, 19, 573–589. [Google Scholar] [CrossRef] [PubMed]
  47. Imtiaz, A.; Shimonaka, S.; Uddin, M.N.; Elahi, M.; Ishiguro, K.; Hasegawa, M.; Hattori, N.; Motoi, Y. Selection of Lansoprazole from an FDA-Approved Drug Library to Inhibit the Alzheimer’s Disease Seed-Dependent Formation of Tau Aggregates. Front. Aging Neurosci. 2024, 16, 1368291. [Google Scholar] [CrossRef] [PubMed]
  48. Gustke, N.; Trinczek, B.; Biernat, J.; Mandelkow, E.-M.; Mandelkow, E. Domains of Tau Protein and Interactions with Micro-tubules. Biochemistry 1994, 33, 9511–9522. [Google Scholar] [CrossRef]
  49. Zhou, Y.; Shi, J.; Chu, D.; Hu, W.; Guan, Z.; Gong, C.-X.; Iqbal, K.; Liu, F. Relevance of Phosphorylation and Truncation of Tau to the Etiopathogenesis of Alzheimer’s Disease. Front. Aging Neurosci. 2018, 10, 27. [Google Scholar] [CrossRef]
  50. Chiarini, A.; Armato, U.; Liu, D.; Dal Prà, I. Calcium-Sensing Receptors of Human Neural Cells Play Crucial Roles in Alzheimer’s Disease. Front. Physiol. 2016, 7, 134. [Google Scholar] [CrossRef]
  51. Lin, L.-F.; Jhao, Y.-T.; Chiu, C.-H.; Sun, L.-H.; Chou, T.-K.; Shiue, C.-Y.; Cheng, C.-Y.; Ma, K.-H. Bezafibrate Exerts Neuropro-tective Effects in a Rat Model of Sporadic Alzheimer’s Disease. Pharmaceuticals 2022, 15, 109. [Google Scholar] [CrossRef]
  52. Jiang, T.; Yu, J.-T.; Zhu, X.-C.; Zhang, Q.-Q.; Cao, L.; Wang, H.-F.; Tan, M.-S.; Gao, Q.; Qin, H.; Zhang, Y.-D.; et al. Temsirolimus Attenuates Tauopathy in Vitro and in Vivo by Targeting Tau Hyperphosphorylation and Autophagic Clearance. Neurophar-macology 2014, 85, 121–130. [Google Scholar] [CrossRef]
  53. Jiang, T.; Yu, J.-T.; Zhu, X.-C.; Tan, M.-S.; Wang, H.-F.; Cao, L.; Zhang, Q.-Q.; Shi, J.-Q.; Gao, L.; Qin, H.; et al. Temsirolimus Promotes Autophagic Clearance of Amyloid-β and Provides Protective Effects in Cellular and Animal Models of Alzheimer’s Disease. Pharmacol. Res. 2014, 81, 54–63. [Google Scholar] [CrossRef]
  54. Bansal, S.; Agrawal, M.; Mahendiratta, S.; Kumar, S.; Arora, S.; Joshi, R.; Prajapat, M.; Sarma, P.; Prakash, A.; Chopra, K.; et al. Everolimus: A Potential Therapeutic Agent Targeting PI3K/Akt Pathway in Brain Insulin System Dysfunction and Associated Neurobehavioral Deficits. Fundamemntal Clin. Pharma 2021, 35, 1018–1031. [Google Scholar] [CrossRef] [PubMed]
  55. Cassano, T.; Magini, A.; Giovagnoli, S.; Polchi, A.; Calcagnini, S.; Pace, L.; Lavecchia, M.A.; Scuderi, C.; Bronzuoli, M.R.; Ruggeri, L.; et al. Early Intrathecal Infusion of Everolimus Restores Cognitive Function and Mood in a Murine Model of Alzheimer’s Disease. Exp. Neurol. 2019, 311, 88–105. [Google Scholar] [CrossRef]
  56. Admane, N.; Grover, A. A Neurohypophyseal Hormone Analog Modulates the Amyloid Aggregation of Human Prion Protein. Biophys. J. 2022, 121, 56a. [Google Scholar] [CrossRef]
  57. Liu, M.; Dexheimer, T.; Sui, D.; Hovde, S.; Deng, X.; Kwok, R.; Bochar, D.A.; Kuo, M.-H. Hyperphosphorylated Tau Aggregation and Cytotoxicity Modulators Screen Identified Prescription Drugs Linked to Alzheimer’s Disease and Cognitive Functions. Sci. Rep. 2020, 10, 16551. [Google Scholar] [CrossRef]
  58. Ashour, N.H.; El-Tanbouly, D.M.; El Sayed, N.S.; Khattab, M.M. Roflumilast Ameliorates Cognitive Deficits in a Mouse Model of Amyloidogenesis and Tauopathy: Involvement of Nitric Oxide Status, Aβ Extrusion Transporter ABCB1, and Reversal by PKA Inhibitor H89. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 111, 110366. [Google Scholar] [CrossRef]
  59. Ko, C.-Y.; Xu, J.-H.; Lo, Y.M.; Tu, R.-S.; Wu, J.S.-B.; Huang, W.-C.; Shen, S.-C. Alleviative Effect of Alpha-Lipoic Acid on Cognitive Impairment in High-Fat Diet and Streptozotocin-Induced Type 2 Diabetic Rats. Front. Aging Neurosci. 2021, 13, 774477. [Google Scholar] [CrossRef] [PubMed]
  60. Salehi, B.; Berkay Yılmaz, Y.; Antika, G.; Boyunegmez Tumer, T.; Fawzi Mahomoodally, M.; Lobine, D.; Akram, M.; Riaz, M.; Capanoglu, E.; Sharopov, F.; et al. Insights on the Use of α-Lipoic Acid for Therapeutic Purposes. Biomolecules 2019, 9, 356. [Google Scholar] [CrossRef] [PubMed]
  61. Dos Santos, S.M.; Romeiro, C.F.R.; Rodrigues, C.A.; Cerqueira, A.R.L.; Monteiro, M.C. Mitochondrial Dysfunction and Al-pha-Lipoic Acid: Beneficial or Harmful in Alzheimer’s Disease? Oxidative Med. Cell. Longev. 2019, 2019, 1–14. [Google Scholar] [CrossRef]
  62. Khan, H.; Singh, T.G.; Dahiya, R.S.; Abdel-Daim, M.M. α-Lipoic Acid, an Organosulfur Biomolecule a Novel Therapeutic Agent for Neurodegenerative Disorders: An Mechanistic Perspective. Neurochem. Res. 2022, 47, 1853–1864. [Google Scholar] [CrossRef]
  63. Pei, X.; Hu, F.; Luo, F.; Huang, X.; Li, X.; Xing, S.; Long, D. The Neuroprotective Effects of Alpha-lipoic Acid on an Experimental Model of Alzheimer’s Disease in PC12 Cells. J. Appl. Toxicol. 2022, 42, 285–294. [Google Scholar] [CrossRef] [PubMed]
  64. Pei, X.; Hu, F.; Hu, Z.; Luo, F.; Li, X.; Xing, S.; Sun, L.; Long, D. Neuroprotective Effect of α-Lipoic Acid against Aβ25–35-Induced Damage in BV2 Cells. Molecules 2023, 28, 1168. [Google Scholar] [CrossRef]
  65. Zara, S.; De Colli, M.; Rapino, M.; Pacella, S.; Nasuti, C.; Sozio, P.; Di Stefano, A.; Cataldi, A. Ibuprofen and Lipoic Acid Conjugate Neuroprotective Activity Is Mediated by Ngb/Akt Intracellular Signaling Pathway in Alzheimer’s Disease Rat Model. Gerontology 2013, 59, 250–260. [Google Scholar] [CrossRef] [PubMed]
  66. Zarini-Gakiye, E.; Vaezi, G.; Parivar, K.; Sanadgol, N. Age and Dose-Dependent Effects of Alpha-Lipoic Acid on Human Mi-crotubule- Associated Protein Tau-Induced Endoplasmic Reticulum Unfolded Protein Response: Implications for Alzheimer’s Disease. CNSNDDT 2021, 20, 451–464. [Google Scholar] [CrossRef]
  67. Zarini-Gakiye, E.; Sanadgol, N.; Parivar, K.; Vaezi, G. Alpha-lipoic Acid Ameliorates Tauopathy-induced Oxidative Stress, Apoptosis, and Behavioral Deficits through the Balance of DIAP1/DrICE Ratio and Redox Homeostasis: Age Is a Determinant Factor. Metab. Brain Dis. 2021, 36, 669–683. [Google Scholar] [CrossRef] [PubMed]
  68. Zhang, Y.-H.; Wang, D.-W.; Xu, S.-F.; Zhang, S.; Fan, Y.-G.; Yang, Y.-Y.; Guo, S.-Q.; Wang, S.; Guo, T.; Wang, Z.-Y.; et al. α-Lipoic Acid Improves Abnormal Behavior by Mitigation of Oxidative Stress, Inflammation, Ferroptosis, and Tauopathy in P301S Tau Transgenic Mice. Redox Biol. 2018, 14, 535–548. [Google Scholar] [CrossRef]
  69. Yao, M.; Rosario, E.R.; Soper, J.C.; Pike, C.J. Androgens Regulate Tau Phosphorylation Through Phosphatidylinositol 3-Kinase–Protein Kinase B–Glycogen Synthase Kinase 3β Signaling. Neuroscience 2025, 568, 503–518. [Google Scholar] [CrossRef]
  70. Duggal, P.; Jadaun, K.S.; Siqqiqui, E.M.; Mehan, S. Investigation of Low Dose Cabazitaxel Potential as Microtubule Stabilizer in Experimental Model of Alzheimer’s Disease: Restoring Neuronal Cytoskeleton. Curr. Alzheimer Res. 2020, 17, 601–615. [Google Scholar] [CrossRef]
  71. Santos, D.B.; Peres, K.C.; Ribeiro, R.P.; Colle, D.; dos Santos, A.A.; Moreira, E.L.G.; Souza, D.O.G.; Figueiredo, C.P.; Farina, M. Probucol, a Lipid-Lowering Drug, Prevents Cognitive and Hippocampal Synaptic Impairments Induced by Amyloid β Peptide in Mice. Exp. Neurol. 2012, 233, 767–775. [Google Scholar] [CrossRef]
  72. Alvarez-de-la-Rosa, M.; Silva, I.; Nilsen, J.; Pérez, M.M.; García-Segura, L.M.; Avila, J.; Naftolin, F. Estradiol Prevents Neural Tau Hyperphosphorylation Characteristic of Alzheimer’s Disease. Ann. Acad. Sci. 2005, 1052, 210–224. [Google Scholar] [CrossRef]
  73. McCormick, A.V.; Wheeler, J.M.; Guthrie, C.R.; Liachko, N.F.; Kraemer, B.C. Dopamine D2 Receptor Antagonism Suppresses Tau Aggregation and Neurotoxicity. Biol. Psychiatry 2013, 73, 464–471. [Google Scholar] [CrossRef]
  74. Majeed, J.; Sabbagh, M.N.; Kang, M.H.; Lawrence, J.J.; Pruitt, K.; Bacus, S.; Reyna, E.; Brown, M.; Decourt, B. Cancer Drugs with High Repositioning Potential for Alzheimer’s Disease. Expert. Opin. Emerg. Drugs 2023, 28, 311–332. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, J.; Zhao, Z.; Lin, E.; Zhao, W.; Qian, X.; Freire, D.; Bilski, A.E.; Cheng, A.; Vempati, P.; Ho, L.; et al. Unintended Effects of Cardiovascular Drugs on the Pathogenesis of Alzheimer’s Disease. PLoS ONE 2013, 8, e65232. [Google Scholar] [CrossRef] [PubMed]
  76. Samir, S.M.; Hassan, H.M.; Elmowafy, R.; ElNashar, E.M.; Alghamdi, M.A.; AlSheikh, M.H.; Al-Zahrani, N.S.; Alasiri, F.M.; Elhadidy, M.G. Neuroprotective Effect of Ranolazine Improves Behavioral Discrepancies in a Rat Model of Scopolamine-Induced Dementia. Front. Neurosci. 2024, 17, 1267675. [Google Scholar] [CrossRef] [PubMed]
  77. Lopez, A.; Siddiqi, F.H.; Villeneuve, J.; Ureshino, R.P.; Jeon, H.-Y.; Koulousakis, P.; Keeling, S.; McEwan, W.A.; Fleming, A.; Rubinsztein, D.C. Carbonic Anhydrase Inhibition Ameliorates Tau Toxicity via Enhanced Tau Secretion. Nat. Chem. Biol. 2025, 21, 577–587. [Google Scholar] [CrossRef]
  78. Yao, J.; Chen, S.R.W. R-Carvedilol, a Potential New Therapy for Alzheimer’s Disease. Front. Pharmacol. 2022, 13, 1062495. [Google Scholar] [CrossRef]
  79. Arya, U.; Dwivedi, H.; Subramaniam, J.R. Reserpine Ameliorates Aβ Toxicity in the Alzheimer’s Disease Model in Caenorhabditis Elegans. Exp. Gerontol. 2009, 44, 462–466. [Google Scholar] [CrossRef]
  80. Vasantharaja, R.; Kumar, A.; Kumar, A.; Subramaniam, J.R. Reserpine Improves Working Memory. J. Behav. Brain Sci. 2016, 6, 107–112. [Google Scholar] [CrossRef]
  81. Hongpaisan, J.; Sun, M.-K.; Alkon, D.L. PKC ε Activation Prevents Synaptic Loss, Aβ Elevation, and Cognitive Deficits in Alz-heimer’s Disease Transgenic Mice. J. Neurosci. 2011, 31, 630–643. [Google Scholar] [CrossRef] [PubMed]
  82. Schrott, L.M.; Jackson, K.; Yi, P.; Dietz, F.; Johnson, G.S.; Basting, T.F.; Purdum, G.; Tyler, T.; Rios, J.D.; Castor, T.P.; et al. Acute Oral Bryostatin-1 Administration Improves Learning Deficits in the APP/PS1 Transgenic Mouse Model of Alzheimer’s Disease. CAR 2015, 12, 22–31. [Google Scholar] [CrossRef]
  83. Zhu, R.; Jiang, G.; Tang, W.; Zhao, X.; Chen, F.; Zhang, X.; Ye, N. Aporphines: A Privileged Scaffold in CNS Drug Discovery. Eur. J. Med. Chem. 2023, 256, 115414. [Google Scholar] [CrossRef] [PubMed]
  84. Fontes Barbosa, M.; Benatti Justino, A.; Machado Martins, M.; Roberta Anacleto Belaz, K.; Barbosa Ferreira, F.; Junio de Oliveira, R.; Danuello, A.; Salmen Espindola, F.; Pivatto, M. Cholinesterase Inhibitors Assessment of Aporphine Alkaloids from Annona Crassiflora and Molecular Docking Studies. Bioorg. Chem. 2022, 120, 105593. [Google Scholar] [CrossRef] [PubMed]
  85. Nabavi, S.M.; Uriarte, E.; Rastrelli, L.; Sobarzo-Sánchez, E. Aporphines and Alzheimer’s Disease: Towards a Medical Approach Facing the Future. Curr. Med. Chem. 2019, 26, 3253–3259. [Google Scholar] [CrossRef] [PubMed]
  86. Sichaem, J.; Tip-pyang, S.; Lugsanangarm, K. Bioactive Aporphine Alkaloids from the Roots of Artabotrys Spinosus: Cholines-terase Inhibitory Activity and Molecular Docking Studies. Nat. Prod. Commun. 2018, 13, 1934578X1801301011. [Google Scholar] [CrossRef]
  87. Zhang, J.; Chen, L.; Sun, J. Oxoisoaporphine Alkaloids: Prospective Anti-Alzheimer’s Disease, Anticancer, and Antidepressant Agents. Chem. Med. Chem. 2018, 13, 1262–1274. [Google Scholar] [CrossRef]
Scheme 1. Methodology workflow. A(F)—value of the ISM amplitude at the frequency F. Amax—maximum amplitude value among all frequencies.
Scheme 1. Methodology workflow. A(F)—value of the ISM amplitude at the frequency F. Amax—maximum amplitude value among all frequencies.
Mps 08 00150 sch001
Figure 1. Structures of DrugBank compounds directly binding to the tau protein.
Figure 1. Structures of DrugBank compounds directly binding to the tau protein.
Mps 08 00150 g001
Figure 2. ISM spectrum of the tau protein.
Figure 2. ISM spectrum of the tau protein.
Mps 08 00150 g002
Figure 3. Cross-spectrum of all five tau-protein-targeting drugs from DrugBank.
Figure 3. Cross-spectrum of all five tau-protein-targeting drugs from DrugBank.
Mps 08 00150 g003
Figure 4. Cross-spectrum of all mammal tau proteins.
Figure 4. Cross-spectrum of all mammal tau proteins.
Mps 08 00150 g004
Figure 5. The domains in the tau protein corresponding to CS frequencies with the ligands.
Figure 5. The domains in the tau protein corresponding to CS frequencies with the ligands.
Mps 08 00150 g005
Figure 6. Distribution of DrugBank hit compounds at different ISM frequencies.
Figure 6. Distribution of DrugBank hit compounds at different ISM frequencies.
Mps 08 00150 g006
Figure 7. Structure of Bryostatin 14.
Figure 7. Structure of Bryostatin 14.
Mps 08 00150 g007
Figure 8. Correlation between S/N and Drug Score of COCONUT compounds. The candidates are colored according to k-means clusters. Red: Cluster 1; Yellow: Cluster 2; Green: Cluster 3; Light Blue: Cluster 4; Dark Blue: Cluster 5.
Figure 8. Correlation between S/N and Drug Score of COCONUT compounds. The candidates are colored according to k-means clusters. Red: Cluster 1; Yellow: Cluster 2; Green: Cluster 3; Light Blue: Cluster 4; Dark Blue: Cluster 5.
Mps 08 00150 g008
Figure 9. Corresponding amino acid residues in the alpha-synuclein, p53 and adrenergic receptor, at their CS frequencies with the ligands.
Figure 9. Corresponding amino acid residues in the alpha-synuclein, p53 and adrenergic receptor, at their CS frequencies with the ligands.
Mps 08 00150 g009
Figure 10. Cross-spectrum of alpha-synuclein and all three targeting ligands.
Figure 10. Cross-spectrum of alpha-synuclein and all three targeting ligands.
Mps 08 00150 g010
Figure 11. CWT plot of alpha-synuclein at all ISM frequencies. The color scale of CWT coefficients is incorporated in the figure.
Figure 11. CWT plot of alpha-synuclein at all ISM frequencies. The color scale of CWT coefficients is incorporated in the figure.
Mps 08 00150 g011
Figure 12. CWT heatmap of alpha-synuclein at ISM frequency F(0.105).
Figure 12. CWT heatmap of alpha-synuclein at ISM frequency F(0.105).
Mps 08 00150 g012
Table 1. Tau-interacting compounds from the DrugBank, with corresponding frequencies from CS with the tau protein.
Table 1. Tau-interacting compounds from the DrugBank, with corresponding frequencies from CS with the tau protein.
DrugBank CompoundNameCS with tau FrequenciesAmplitudeS/NCorresponding Domain in the tau ProteinLiterature Binding Region
DB00637Astemizole0.3420.995817.353269–525386–391
DB01248Docetaxel0.1671.784720.02362–318β-tubulin
DB14914Flortaucipir F-180.0800.108169.0225427–683R3–R4 386–391
DB00448Lansoprazole0.4350.6551412.299305–561R3–R4 386–391
DB01229Paclitaxel0.1941.191714.08923–279β-tubulin
Table 2. The list of identified DrugBank compounds from the tau protein ISM spectrum.
Table 2. The list of identified DrugBank compounds from the tau protein ISM spectrum.
NoIDNameAmplitudeS/NFrequencyEffect on tau Protein/AD
1DB01012Cinacalcet0.4048113.452780.080Indirect on tau phosphorylation
2DB01393Bezafibrate0.327239.67330.080Reduces Aβ and tau pathology
3DB06287Temsirolimus1.1483328.351910.167Reducing tau hyperphosphorylation
4DB01590Everolimus3.2377620.748360.167Reducing tau hyperphosphorylation
5DB00035Desmopressin2.3031320.559080.167Could influence Aβ/tau cross-interactions
6DB01130Prednicarbate1.3460918.589360.167Potential tau aggregation modulator
7DB01656Roflumilast17.850850.059620.167Ameliorates cognitive deficits in tauopathy models
8DB00166Lipoic acid17.14430.040870.167Reduces tauopathy
9DB01420Testosterone Propionate1.0059522.219790.194Hyperphosphorylation of tau
10DB06772Cabazitaxel2.4751522.126930.194Microtubule stabilization
11DB01599Probucol0.5027119.558320.194Reduce amyloid deposition
12DB08866Estradiol valerate/Dienogest0.9107219.028540.194Prevents tau hyperphosphorylation
13DB00850Perphenazine0.7194814.657170.333Lower the levels of insoluble tau.
14DB06699Degarelix2.0554113.785090.333Hormone modulation may influence neurodegeneration
15DB00883Isosorbide Dinitrate0.8745322.008890.342Nitric oxide modulation (could influence neurodegeneration)
16DB00243Ranolazine1.4800223.25340.435Reduces oxidative stress, lacks tau-specific evidence
17DB00423Methocarbamol0.8044121.587010.435Promoting tau clearance
18DB01136Carvedilol1.0770721.143590.435May reduce Aβ and tau toxicity
19DB00206Reserpine2.1540119.969370.435Reduces Aβ toxicity
Table 3. The top ten compounds from the COCONUT database, at all frequencies, ranked by Total Score. Bryostatin-14 is printed in bold.
Table 3. The top ten compounds from the COCONUT database, at all frequencies, ranked by Total Score. Bryostatin-14 is printed in bold.
Compound IDAmplitudeS/NDrug ScoreTotal ScoreF
CNP0504067.02.693127.51460.423611.65480.080
CNP0126636.12.046222.20790.38718.5975
CNP0560502.00.944720.84650.40408.4210
CNP0195295.11.737422.26900.37348.3160
CNP0532732.00.591616.87570.46197.7952
CNP0581434.00.914018.88450.41167.7720
CNP0111317.11.083521.70100.35037.6024
CNP0154283.11.774819.10530.39377.5220
CNP0285595.10.911221.77440.33307.2507
CNP0511033.00.780421.42970.33237.1204
CNP0266316.14.279033.72200.384912.98060.167
CNP0168057.14.989535.85600.345012.3714
CNP0427543.13.508037.84520.318112.0369
CNP0135438.13.023439.19000.300211.7653
CNP0327834.12.501436.20930.322411.6753
CNP0297394.13.159331.30390.355811.1366
CNP0359990.13.048834.43670.316410.8969
CNP0449680.16.379530.72000.354410.8859
CNP0072358.11.562529.16540.373210.8849
CNP0280000.14.705230.87410.351610.8550
CNP0267855.17.152754.76950.341618.71030.194
CNP0115161.15.963948.03600.338916.2787
CNP0271940.16.007847.16670.332815.6975
CNP0144759.13.828832.95080.464015.2877
CNP0399889.15.285942.04290.338914.2478
CNP0271195.15.097839.06680.332813.0018
CNP0206347.15.216127.66650.466012.8917
CNP0075233.14.161335.81590.334811.9926
CNP0199404.12.089647.56280.249711.8770
CNP0337940.13.917429.68840.393011.6670
CNP0425508.18.053941.71910.386116.10710.333
CNP0426456.17.355640.40950.382415.4512
CNP0580557.079.8827108.70100.120513.0938
CNP0492610.16.660145.59390.265512.1062
CNP0574550.14.460724.83690.482011.9716
CNP0493035.14.374824.46050.482011.7901
CNP0598400.05.283429.81020.391611.6749
CNP0571478.15.534038.77410.265510.2954
CNP0491847.15.625439.18780.255610.0162
CNP0357360.07.969139.01200.25569.9713
CNP0285895.18.689943.58420.387716.89880.341
CNP0313376.18.345042.95290.370415.9113
CNP0578185.16.396232.05140.443714.2199
CNP0291861.16.257939.10640.350113.6928
CNP0538593.13.968333.60900.403913.5734
CNP0180487.05.745136.78960.362813.3468
CNP0525297.16.129037.81860.352113.3144
CNP0423521.15.303245.47800.281112.7837
CNP0549106.17.123837.67060.334112.5872
CNP0319138.14.855033.64160.367112.3481
CNP0551487.116.057980.20870.238919.16400.435
CNP0199424.06.898548.91950.312115.2669
CNP0509389.23.925334.60860.434815.0463
CNP0105199.115.390473.53800.199114.6407
CNP0417346.04.444436.20800.396014.3374
CNP0048849.126.472780.32430.174213.9922
CNP0061932.12.424238.15900.365713.9535
CNP0151916.02.410737.60990.349213.1339
CNP0078724.120.898853.48320.244813.0907
CNP0429159.11.927728.38670.451012.8016
Table 4. The top compounds from the COCONUT database per cluster, ranked by Total Score 2. Bryostatin-14 and CNP0151916.0 are printed in bold.
Table 4. The top compounds from the COCONUT database per cluster, ranked by Total Score 2. Bryostatin-14 and CNP0151916.0 are printed in bold.
Compound IDS/NDrug ScoreDistance to ClusterFTotal Score 2 (S/N/Distance to Centroid)Cluster
CNP0151916.037.60990.349223.17680.4351.62271
CNP0327834.136.20930.322453.61440.1670.6754
CNP0072358.129.16540.373244.14540.1670.6607
CNP0504067.027.51460.423654.13430.080.5083
CNP0199404.147.56280.2497110.95280.1940.4287
CNP0061932.138.15900.3657101.69110.4350.3752
CNP0427543.137.84520.3181103.77780.1670.3647
CNP0135438.139.19000.3002132.43140.1670.2959
CNP0511033.021.42970.332375.76210.080.2829
CNP0560502.020.84650.404079.23960.080.2631
CNP0111317.121.70100.350397.89610.080.2217
CNP0359990.134.43670.3164164.06050.1670.2099
CNP0581434.018.88450.411694.38670.080.2001
CNP0285595.121.77440.3330110.41450.080.1972
CNP0532732.016.87570.4619105.89510.080.1594
CNP0195295.122.26900.3734173.70490.080.1282
CNP0337940.129.68840.393015.28770.1941.94202
CNP0199424.048.91950.312144.92650.4351.0889
CNP0574550.124.83690.482026.61040.3330.9334
CNP0493035.124.46050.482026.32540.3330.9292
CNP0266316.133.72200.384944.73050.1670.7539
CNP0126636.122.20790.387135.90000.080.6186
CNP0105199.173.53800.1991140.42450.4350.5237
CNP0168057.135.85600.345078.07690.1670.4592
CNP0429159.128.38670.451064.16860.4350.4424
CNP0154283.119.10530.393746.26990.080.4129
CNP0423521.145.47800.2811115.18180.3410.3948
CNP0417346.036.20800.3960107.34360.4350.3373
CNP0297394.131.30390.355895.05480.1670.3293
CNP0180487.036.78960.362813.95200.3412.63693
CNP0313376.142.95290.370420.42500.3412.1030
CNP0525297.137.81860.352134.16820.3411.1068
CNP0319138.133.64160.367144.94900.3410.7484
CNP0291861.139.10640.350166.29430.3410.5899
CNP0285895.143.58420.387779.08450.3410.5511
CNP0598400.029.81020.391657.84190.3330.5154
CNP0280000.130.87410.351677.42120.1670.3988
CNP0538593.133.60900.403999.86430.3410.3365
CNP0206347.127.66650.466087.91750.1940.3147
CNP0115161.148.03600.338964.06380.1940.74984
CNP0271940.147.16670.332863.06340.1940.7479
CNP0399889.142.04290.338964.24650.1940.6544
CNP0271195.139.06680.332862.72610.1940.6228
CNP0267855.154.76950.3416112.74270.1940.4858
CNP0075233.135.81590.334878.41680.1940.4567
CNP0580557.0108.70100.1205266.91200.3330.4073
CNP0426456.140.40950.3824151.00780.3330.2676
CNP0425508.141.71910.3861166.11880.3330.2511
CNP0357360.039.01200.2556163.79660.3330.2382
CNP0078724.153.48320.2448356.33950.4350.1501
CNP0144759.132.95080.4640224.42410.1940.1468
CNP0551487.180.20870.2389134.29480.4350.59735
CNP0048849.180.32430.1742137.08050.4350.5860
CNP0509389.234.60860.434879.41400.4350.4358
CNP0492610.145.59390.2655121.36310.3330.3757
CNP0491847.139.18780.2556106.63580.3330.3675
CNP0549106.137.67060.3341108.18710.3410.3482
CNP0578185.132.05140.443795.46390.3410.3357
CNP0571478.138.77410.2655121.36450.3330.3195
CNP0449680.130.72000.3544102.71580.1670.2991
Table 5. The key residues for IDP–ligand interactions were compared from MD simulations with the Martini-IDP forcefield and the ISM-SM method. The residues most approximately identified in both methods are in bold.
Table 5. The key residues for IDP–ligand interactions were compared from MD simulations with the Martini-IDP forcefield and the ISM-SM method. The residues most approximately identified in both methods are in bold.
SystemMartini-IDPISM-SM Region
alpha-synuclein- fasudil3, 38, 93, 115, 124, 127, 1351, 2, 7, 23, 24, 27, 28, 66, 67, 68, 69,70, 71, 104, 108, 109 (Slide window width 17)
p53—Ligand 10502321, 38, 119, 184, 258, 305, 329, 361 (Slide window width 33)
AR—EPI-002396, 405, 406, 432, 433, 437, 438177, 289, 407, 563, 689, 765
(Slide window width 8)
Table 6. Top peaks for each alpha-synuclein–ligand interaction, for all three ligands’ common interaction and corresponding protein domains from slide window analysis.
Table 6. Top peaks for each alpha-synuclein–ligand interaction, for all three ligands’ common interaction and corresponding protein domains from slide window analysis.
Protein–Ligand ComplexPeakFAS/NISM-SM Region
Ligand4710.0540.077211.1842–106
20.1050.06158.91474–132
30.2260.04386.347470–134
Fasudil10.1050.097212.3594–132
20.0310.07209.158757–121
30.0930.06678.47967–71
Ligand2310.3000.03427.499357–121
20.3510.02986.544571–135
30.1050.02876.29844–132
All three ligands10.1050.000348.29844–132
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Senćanski, M. New Approach for Targeting Small-Molecule Candidates for Intrinsically Disordered Proteins. Methods Protoc. 2025, 8, 150. https://doi.org/10.3390/mps8060150

AMA Style

Senćanski M. New Approach for Targeting Small-Molecule Candidates for Intrinsically Disordered Proteins. Methods and Protocols. 2025; 8(6):150. https://doi.org/10.3390/mps8060150

Chicago/Turabian Style

Senćanski, Milan. 2025. "New Approach for Targeting Small-Molecule Candidates for Intrinsically Disordered Proteins" Methods and Protocols 8, no. 6: 150. https://doi.org/10.3390/mps8060150

APA Style

Senćanski, M. (2025). New Approach for Targeting Small-Molecule Candidates for Intrinsically Disordered Proteins. Methods and Protocols, 8(6), 150. https://doi.org/10.3390/mps8060150

Article Metrics

Back to TopTop