Abstract
Intrinsically disordered proteins (IDPs) are now well-recognized as drug targets. Identifying drug-interacting residues (DIRs) is valuable for both optimizing compounds and elucidating the mechanism of action. Currently, NMR chemical shift perturbation and all-atom molecular dynamics (MD) simulations are the primary tools for this purpose. Here we present DIRseq, a fast method for predicting DIRs from the amino-acid sequence. All residues contribute to the propensity of a particular residue to be drug-interacting; the contributing factor of each residue has an amplitude that is determined by its amino-acid type and attenuates with increasing sequence distance from the particular residue. DIRseq predictions match well with DIRs identified by NMR chemical shift perturbation and other methods, including residues L22WK24 and Q52WFT55 in the tumor repressor protein p53. These successes augur well for deciphering the sequence code for IDP-drug binding. DIRseq is available as a web server at https://zhougroup-uic.github.io/DIRseq/ and has many applications, such as virtual screening against IDPs and designing IDP fragments for in-depth NMR and MD studies.
Introduction
Intrinsically disordered proteins (IDPs) are now recognized as important drug targets 1-4. For structured protein targets, a crucial step is identifying the drug-binding pocket. Although an IDP can be locked into a specific conformation by a drug molecule in rare cases 5, the prevailing scenario is that the protein remains disordered upon drug binding. Consequently, the IDP-drug complex typically samples a vast conformational space, and the drug molecule only exhibits preferences, rather than exclusiveness, for interacting with subsets of residues. Such drug-interacting residues (DIRs), akin to binding pockets in structured proteins, are key to optimizing compounds and elucidating the mechanism of action.
NMR chemical shift perturbation (CSP) is the best experimental method for identifying DIRs and has been applied to many IDPs 6-23. Aromatic residues are frequently among DIRs 7,10,12,17,18,20,22. This is understandable as drug molecules typically are rich in aromatic rings (Figure 1 and Table S1), which can form π-π interactions with aromatic residues. As recently pointed out by Heller et al. 23, CSPs of IDPs elicited by drug binding can be small enough to fall within the spectral resolution of NMR spectroscopy, therefore making it difficult to unequivocally identify DIRs. The small magnitude of CSPs may arise because the drug does not have a strong preference for interacting with any residues. Another scenario may be that drug binding induces a conformational shift such as secondary structure formation or even partial folding 11,16,20, so CSPs spread from the directly interacting residues to the rest of the IDP, adding to the difficulty in identifying DIRs.

Four IDPs and drugs that bind to them.
(a) p27, p21, and SJ403. (b) p53 and EGCG. (c) -synuclein and Fasudil. For each IDP, the DIRseq propensities are rendered by a color spectrum from yellow for low values to red for high values. Predicted DIRs are shown with sidechains rendered in stick.
All-atom molecular dynamics (MD) simulations have presented atomic details in many IDP-drug systems 17,18,20,22,24-29. These simulation studies have highlighted the frequent engagement of drug molecules with aromatic residues, particularly in π-π interactions 17,18,20,22,26-29. MD simulations also revealed the emergence of a compact sub-population of the N-terminal disordered region of the tumor repressor protein p53 upon binding the drug epigallocatechin-3-gallate (EGCG) 18 and reduced solvent exposure of hydrophobic residues in the 42-residue amyloid-β peptide (Aβ42) upon binding the drug 10074-G5 25. However, MD simulations of IDPs still suffer from the perennial issues of inaccurate force fields and insufficient sampling, which are exacerbated when drug molecules are included. For example, two replicate simulations may identify different DIRs 30.
Here we present a sequence-based method, DIRseq, as a complement to NMR CSP and MD simulations. This method was motivated by our observation that DIRs seem to overlap with residues exhibiting elevated transverse relaxation rates (R2) 31,32, as exemplified by the C-terminal 20 residues of α-synuclein 20. Elevated R2 in IDPs is caused by either local inter-residue interactions or residual secondary structure 33. Based on this understanding, we developed a sequence-based method, SeqDYN, to predict R2 of IDPs 31. As suggested previously 32, the propensities of residues to form intramolecular interactions and therefore elevate R2 should be similar to those for forming intermolecular interactions with drug molecules. DIRseq is therefore an adaptation of SeqDYN. DIRseq predictions match well with DIRs identified by NMR chemical shift perturbation and other methods, including residues L22WK24 and Q52WFT55 in p53 and C-terminal residues in α-synuclein. DIRseq is available as a web server at https://zhougroup-uic.github.io/DIRseq/ and has many applications.
Results
Retooling of SeqDYN into DIRseq
SeqDYN 31 predicts the R2 value of residue n by accumulating a contributing factor f(i; n) from each residue i:
where N is the total number of residues in the IDP and ϒ is a uniform scale parameter. The contributing factor f(i; n) has an amplitude q(i) that is determined by the amino-acid type of residue i and attenuates with increasing sequence distance, s = |i − n|, from residue n:
The 20 q parameters (one for each amino-acid type) and b were trained on the measured R2 values for 45 IDPs. The original q values (Figure S1a) show that aromatic (Trp, Tyr, His, and Phe), Arg, and long aliphatic (Ile and Leu) amino acids are interaction-prone and tend to elevate R2. We transformed SeqDYN into DIRseq by implementing four changes.
First, we reassigned four q parameters. We lowered the q values of the three long aliphatic amino acids, Leu, Ile, and Met, to the q value of a short aliphatic amino acid, Val, because long aliphatic amino acids primarily participate in hydrophobic interactions, which may be less important for stabilizing the binding of a small molecule in sites largely exposed to water. At the same time, we raised the q value of Asp to be the same as that of Glu, to increase the role of Asp’s electrostatic interactions. The modified set of q parameters is displayed in Figure S1b.
Second, the original b value, 0.0316, corresponds to a correlation half-length, b−1/2, of 5.6 residues. Given the small size of drug molecules, we increased the b value to 0.3, corresponding to a correlation half-length of 1.8 residues. That is, we expect a drug molecule to interact with 3 to 4 residues at a time. Third, the SeqDYN-predicted R2 profile, capturing experimental observations 34, falls off at both termini, because no residues beyond the termini are present to provide a contributing factor to R2. However, whereas intra-IDP interactions experience such a terminal effect, IDP-drug interactions do not. To eliminate the terminal effect, we padded the original IDP sequence by a stretch of 12 Gln residues at each terminus. Gln was selected because its q value is at the middle of the 20 q values (Figure S1b).
Lastly, we converted high and low R2 values into high and low DIR propensities, respectively, using a sigmoid function
where the midpoint R2th and width R2wd of the transition region are determined by the mean (m) and standard deviation (SD) of the R2 values over the entire sequence. Specifically,
We chose s1 to be 1.5 and s2 to be 14.0. On the DIRseq web server, users can either keep these default values for b, s1, and s2, or enter values of their choice. The values of p range from 0 to 100; p = 50 when R2 is at the “threshold” value R2th. Residues with p ≥ 50 are predicted to be DIR, but an isolated residue with p ≥ 50 is discarded, as it can be reasonably expected that interactions with at least two consecutive (or nearby) residues are needed to generate sufficient drug binding stability.
Detailed assessment of DIRseq on four IDPs
DIRs in four IDPs or intrinsically disordered regions (IDRs) have been thoroughly characterized by NMR CSP. In Figure 1 (left images), we show these IDPs in a single conformation, with DIRseq propensities rendered in a color spectrum (from low in yellow to high in red); predicted DIRs (i.e., those with p ≥ 50) are shown with sidechains in stick. Drug molecules that bind to these IDPs are also displayed in Figure 1 (right images). We display NMR CSPs as blue bars and DIRseq propensities as a red curve in Figure 2. Unless otherwise noted, we use m + 1.5 SD as the threshold for identifying DIRs, for both CSP and DIRseq; this threshold is indicated by a horizontal dashed line in Figure 2. Experimentally identified DIRs are further indicated by cyan shading. Below we present a detailed comparison between CSP and DIRseq.

Comparison of DIRseq propensities with NMR CSPs.
(a) p27. (b) p21. (c) p53. (d) -synuclein. CSPs are displayed as blue bars and in units of ppb; CSP-identified DIRs are indicated by cyan shading. DIRseq predictions are shown as red curves. The ordinate scales are chosen so that the m + 1.5 SD threshold for CSP is at the same height as the 50% threshold for DIRseq, indicated by a horizontal dashed line.
The kinase inhibitory domain (residues 22-105) of the cell cycle regulator p27 harbors three aromatic-centered motifs that interact with a group of compounds represented by SJ403 (Figure 1a), as found by Iconaru et al. 10. These motifs, W60N61, E75WQ77, and Y88Y89 (Figure 2a, cyan shading), are correctly predicted by DIRseq (Figure 2a, portions of the red curve above the 50% threshold). Specifically, the predicted DIRs are R58KWN61, Y74EWQ77, and Y88Y89 (Figure 1a, left image). Moreover, the DIRseq propensity profile matches well the CSP profile along the sequence (Figure 2a). The two parameters show a very strong correlation, with a Pearson correlation coefficient (r) of 0.82 (Figure S2a).
Iconaru et al. also studied SJ403 binding to a related cell cycle regulator p21. Their CSP data identified the first two corresponding motifs, W49N50 and F63A64, but not the third as DIRs (Figure 2b, cyan shading). DIRseq correctly predicts both the first two motifs above the 50% threshold and the third motif below the threshold (Figure 2b, red curve; Figure 1b, left image). As noted by Iconaru et al., the p21 counterpart to the p27 third motif F87YY89 is L76YL78; the two Leu residues in p21 are no match for F87 and Y89 in p27. For the DIRseq method, Leu has a lower q value than Tyr and Phe (Figure S1b). CSP and DIRseq propensity show a strong correlation (r = 0.66; Figure S2b).
NMR CSP revealed two motifs, W23K24 and D49IEQWFT55, in the N-terminal region of p53 as interacting with EGCG 18 (Figure 2c, cyan shading). MD simulations supported these drug-binding sites. DIRseq again correctly predicts these two motifs (Figure 2c, red curve). Specifically, the predicted DIR motifs are L22WK24 and E51QWFT55 (Figure 1c, left image). Here also CSP and DIRseq propensity show a strong correlation (r = 0.67; Figure S2c).
Using the m + 1.5 SD threshold, CSPs of Robustelli et al. 20 select residues A124YEMPS129 and Y133QDYE137 of α-synuclein as interacting with the drug Fasudil (Figure 2d, cyan shading). These two C-terminal motifs are also identified by DIRseq, with residues A124YE126 and Y133QDYEP138 above the 50% threshold (Figure 2d, red curve; Figure 1d, left image). There is a moderate correlation (r = 0.51; Figure S2d) between CSP and DIRseq propensity. Robustelli et al. further characterized the IDP-drug interactions by all-atom simulations, using both full-length α-synuclein and a C-terminal fragment (residues 121-140). Further simulations of the C-terminal fragment binding with additional compounds led to a compound known as Ligand 47 with a somewhat higher affinity than Fasudil. The CSP profiles elicited by Fasudil and Ligand 47 are similar, though with larger amplitudes around Y125 and Y136 by the latter; their correlation coefficient is 0.78 (Figure S3a). Because DIRseq does not consider any information about the drug molecule, this correlation coefficient between CSPs elicited by two different compounds might be viewed as an upper bound of what can be achieved for the correlation between CSP and DIRseq propensity.
DIRseq as a complement to CSP for assigning DIRs
After assessing the achievable accuracy of DIRseq, we now consider an application: its combination with CSP to make robust assignments of DIRs. We note that the above CSP-identified drug-interacting motifs are all anchored on one or more aromatic residues, and this feature likely contributes to the good performance of DIRseq. When a clear “aromatic signal” is not present, CSP or DIRseq alone may not be able to conclusively identify DIRs. However, a consensus identification by CSP and DIRseq may be reliable. MD simulations have played such a complementary role to CSP in several studies 18,20,22,28.
De Mol et al. 11 reported the CSPs of an IDR called AF-1* (residues 142-448) in the androgen receptor elicited by EPI-001 and its stereoisomers including EPI-002; small but reproducible CSPs were found in three subregions, R1 (residues 341-371), R2 (residues 391-414), and R3 (residues 426-446). Conversely, AF-1* caused changes in the 1H NMR spectrum of EPI-001 but the peptides corresponding to R1-R3 did not, suggesting that, rather than separately interacting with the three subregions, EPI-001 induces a partial folding of the three subregions. MD simulations captured the partial folding of the R2-R3 fragment (residues 391-446) induced by EPI-002, and underscored the importance of aromatic residues in drug binding 28. Basu et al. 22 reported the CSPs of the transactivation unit 5 (Tau-5*, residues 336-448) in AF-1* by EPI-001 and a more potent variant, 1aa (Figure 3a and Figure S3b). The contact probabilities of the R2-R3 fragment with 1aa in MD simulations reaffirmed the importance of aromatic residues.

Drug-binding sites identified by combining CSP or mutation data with DIRseq predictions.
(a) Tau-5*. (b) NS5A-D2D3. (c) β2 microglobulin. Display items in panels (a, b) have the same meanings as in Figure 2, except that cyan shading indicates consensus identification; in panel (c), vertical lines indicate mutation sites.
De Mol et al. 11 and Basu et al. 22 were careful not to name any DIRs based on CSPs. In Figure 3a, we compare the 1aa-elicited CSPs and DIRseq propensities of Tau-5*. In agreement with both NMR studies, DIRseq identifies DIRs in the middle of each of R1-R3: R360DYY363, A396WAA399, and W433H434. In addition, the latter two motifs showed the highest drug-contact probabilities in separate MD simulations of the R2-R3 fragment 22,28. For the R2 and R3 subregions, CSPs above the m + 1.5 SD threshold are observed at residues downstream of the DIRseq identifications, so we propose to expand the drug-interacting motifs to A396WAAAAAQ403 and W433HTLF437. The three putative drug-interacting motifs are indicated as cyan shading in Figure 3a.
Heller et al. 23 used 19F transverse relaxation measurements to determine the binding affinity of the disordered domains 2 and 3 of the hepatitis C virus NS5A protein (NS5A-D2D3, residues 247-466) for 5-fluorindole. They also measured 1H-15N CSPs at two ligand concentrations but described them as “nearly undetectable” (Figure 3b). We speculate that the small CSPs may be due to the small size of the ligand, making it difficult to interact with multiple residues simultaneously and thus achieve sufficient binding stability. In any event, CSPs above the m + 1.5 SD threshold were largely isolated (and thus appeared to be random), and there was not much overlap between residues having these above-the-threshold CSPs at the two ligand concentrations. The one exception is a motif around W312, which had above-the-threshold CSPs at both ligand concentrations, and nearby residues A308 and A313 also had above-the-threshold CSPs at one of the ligand concentrations. DIRseq predicts the motif P310AWARPD316 as DIRs. We propose the expanded motif, A308LPAWARPD316, as residues interacting with 5-fluorindole (Figure 3b, cyan shading). DIRseq also predicts two more motifs, E323SWRRPDY330 and R352RRR355, as DIRs, which remain to be tested.
The fibrillation of acid-denatured β2 microglobulin is inhibited by rifamycin SV 35. An aromatic-rich motif, W60SFYLLYYTEF70, was implicated in the nucleation of fibrillation and also involved in ligand binding, as a triple mutation, F62A/Y63A/Y67A, significantly weakened binding. Low intensities of NMR peaks from residues 58-79 (possibly due to the formation of residual structures) prevented the measurement of CSPs. DIRseq predicts D59WSFY63 as DIRs. Combined with the mutational data of Woods et al. 35, we propose the expanded motif, D59WSFYLLYY67, as the major drug-binding site (Figure 3c, cyan shading). DIRseq also predicts an additional motif, K94WDR97, as DIRs.
The aggregation of human islet amyloid polypeptide (hIAPP; 37 residues) is inhibited by the small molecule YX-I-1 21. CSPs elicited by this molecule were small (Figure 4a). In addition, CSPs of short IDPs may not exhibit strong disparities because amino acids may be too well mixed along the sequence or drug binding may induce a conformational shift. We thus reduce the threshold for identifying DIRs to m + 1.0 SD when the number of residues is ≤ 50. With this threshold, three residues are identified by CSP as DIRs: R11 and V17H18. In comparison, DIRseq identifies T9QRLA13 and F15 as DIRs, which partially overlap with the CSP identifications. Combining the two types of data, we propose T9QRLANFLVH18 as the primary drug-binding site (Figure 4a, cyan shading). We note that this motif is also prone to α-helix formation 36.

Drug-binding sites identified by combining CSP or mutation data with DIRseq predictions.
(a) hIAPP. (b) Aβ42. (c) c-Myc. In panel (b), vertical lines indicate residues with prominent CSPs; those accompanied by NMR peak broadening have their vertical lines in dark color. The threshold for identifying DIRs is lowered to m + 1.0 SD.
Ono et al. 8 acquired 1H-15N heteronuclear single quantum coherence spectra of the 42-residue amyloid-β (Aβ42) in the absence and presence of the oligomerization-blocking compound myricetin. CSPs were not calculated, but chemical shift movements were most pronounced at R5, V12H13, K16LVFFAED23, and I31I32 (vertical lines in Figure 4b). In addition, NMR cross peaks suffered broadening upon ligand binding at four of these residues: R5, V12, K16, and V18 (dark vertical lines in Figure 4b), implicating elevated probabilities of ligand interactions. DIRseq predicts E3FRH6 and H13H14 as DIRs. Combined with the NMR data, we propose E3FRH6 and V12HHQKLV18 as the primary ligand-binding sites (Figure 4b, cyan shading). Aβ42 CSPs elicited by several other compounds were also widely distributed over the sequence, such that Iwaya et al. 16 “failed to identify” DIRs, implicating a conformational shift.
Hammoudeh et al. 6 identified three distinct drug binding sites in a C-terminal IDR (residues 363-412) of the oncoprotein c-Myc. These authors measured the CSPs of three overlapping fragments (residues 363-381, 370-409, and 402-412) each in the presence of a single compound and of the full IDR in the presence of all three compounds. Using 20 parts per billion (ppb) as the threshold, Hammoudeh et al. named residues R366RNELKRSFF375, F375ALRDQIPELE385, and Y402ILSVQAE409 as the binding sites for 10074-G5, 10074-A4, and 10058-F4, respectively. We present their CSP data for the full IDR in Figure 4c. Using the m + 1.0 SD threshold, only six residues are identified as DIRs: R367, L370, F374F375, A399, and S405. DIRseq predicts two motifs, E363RQRR367 and K371RSFFA376, that overlap with the first two sites identified by CSP; a third motif, K397KATAY402, that corresponds to the third CSP-identified site has moderate DIR propensities. Combining CSP and DIRseq, we revise the three drug-binding sites to be E363RQRRNELKRSFF375, S373FFALRDQI381, and K397KATAYILS405 (Figure 4c, cyan, olive, and yellow shading, respectively). In addition to 10074-G5, 10074-A4, and 10058-F4, many compounds bind to these three sites 24,37–39.
Discussion
We have presented the first sequence-based method, DIRseq, to predict drug-interacting residues of IDPs. Assessment against NMR CSP demonstrates the accuracy of DIRseq. Drug-binding motifs are anchored on one or more aromatic residues, for forming π-π interactions with drug-like molecules that are rich in aromatic groups. The success of DIRseq comes without any specific information on the drug molecules, suggesting that IDPs may have a relatively simple sequence code for drug binding.
The notion that DIRs may be agnostic to the molecular details of drug compounds is supported by the fact that the same drug can bind to different IDPs. For example, in addition to p53 18 (Figure 2c), the polyphenol EGCG also binds to many other IDPs, including α-synuclein 40, hIAPP 41,42, Aβ40 13 and Aβ42 40, tau 43, and merozoite surface protein 2 44. Likewise, 10074-G5 binds to c-Myc 6 (Figure 4c) but also to Aβ42 25. On the other hand, c-Myc represents a case where different compounds bind to distinct sites on a single IDP 6. A related example is presented by p27, where SJ403 typifies a group of compounds that share the same three binding sites (Figure 2a). Another group of compounds, typified by SJ710, binds only to the third site. Chemically, the presence of nitrogen atoms in the rings of SJ403 enhances its aromaticity and thus strengthens π-π interactions; in addition, the electronegative groups of SJ403 project into different directions, making it less restricted when forming electrostatic interactions (Figure S4). These features may explain why SJ403 can bind to all three sites whereas SJ710 can bind only to the third site, F87YYR90, where three consecutive aromatic residues followed by a basic residue ensure that SJ710 can form both π-π and electrostatic interactions.
We have illustrated the combination of DIRseq with NMR CSP to make robust identifications of drug-binding sites in IDPs. Indeed, CSP, MD simulations, and DIRseq are three orthogonal approaches that have great potential in complementing each other, not only for identifying drug-binding sites but also for elucidating the roles of amino acids, their sequence context, and different types of noncovalent interactions in forming such sites. DIRseq offers fast speed and a simple, direct link between sequence motif and propensity for drug binding. As more data from CSP and MD simulations become available, DIRseq can be tuned to make even better predictions. For example, it may be feasible to develop DIRseq versions specific to different pharmacophores.
Another application of DIRseq is to define IDP fragments for in-depth study by MD simulations, as shorter constructs both enable the use of a smaller simulation box and reduce the size of the conformational space. For example, based on CSP data and initial MD simulations of fill-length of -synuclein, Robustelli et al. 20 chose a 20-residue C-terminal fragment for simulations of binding with additional compounds, leading to the identification of Ligand 47 as a stronger binder than the original Fasudil. Similarly, based on CSP data from longer constructs of the androgen receptor, both Zhu et al. 28 and Basu et al. 22 chose the 56-residue R2-R3 fragment for MD simulations of drug binding. DIRseq can now play a similar role in selecting fragments for MD simulations when CSP data are unavailable. Longer constructs may also present challenges such as resonance assignments to NMR experiments, so well-chosen fragments guided by DIRseq can also benefit NMR studies.
Lastly, virtual screening has been conducted against conformational ensembles of IDPs 17,45; drug-binding sites predicted by DIRseq can be used to guide such screening. As a simple illustration, we present poses of EGCG generated by screening against the two DIRseq-predicted binding sites in p53 in Figure 5. As IDPs sample a vast conformational space, knowledge of the binding site can drastically reduce the computational cost. The subset of conformations that generate high docking scores for a given drug at the known site can also provide insight into the mechanism of drug action.

Poses of p53-bound EGCG generated by docking.
Acknowledgements
This work was supported by National Institutes of Health Grant GM118091.
Additional information
Computational Methods
The sequences of the IDP studied here and the drugs that bind to them are listed in Table S1. All DIRseq predictions were obtained using the web server at https://zhougroup-uic.github.io/DIRseq/. Conformations of IDPs were generated using the TraDES method 46.
Docking of EGCG onto p53 was performed via the SwissDock web server at https://www.swissdock.ch/ 47 utilizing the Autodock Vina docking engine 48. The SMILES string for EGCG from PubChem (https://pubchem.ncbi.nlm.nih.gov/; CID 65064) and several conformations of p53 were used as input. A cubic region (13 to 20 Å in side length) around the center of each DIR was selected for docking.
Funding
National Institute of General Medical Sciences (GM118091)
Additional files
References
- (1)Druggability of Intrinsically Disordered ProteinsIn:
- Felli I. C.
- Pierattelli R.
- (2)Fuzzy Drug Targets: Disordered Proteins in the Drug-Discovery RealmACS Omega 8:9729–9747Google Scholar
- (3)Rational drug design targeting intrinsically disordered proteinsWiley Interdiscip Rev Comput Mol Sci 13:e1685Google Scholar
- (4)How to drug a cloud? Targeting intrinsically disordered proteinsPharmacol Rev 77https://doi.org/10.1124/pharmrev.124.001113Google Scholar
- (5)Chemical inhibition of N-WASP by stabilization of a native autoinhibited conformationNat Struct Mol Biol 11:747–755Google Scholar
- (6)Multiple Independent Binding Sites for Small-Molecule Inhibitors on the Oncoprotein c-MycJ Am Chem Soc 131:7390–7401Google Scholar
- (7)Structural and mechanistic basis behind the inhibitory interaction of PcTS on α-synuclein amyloid fibril formationProc Natl Acad Sci U S A 106:21057–21062Google Scholar
- (8)Phenolic Compounds Prevent Amyloid β-Protein Oligomerization and Synaptic Dysfunction by Site-specific Binding*J Biol Chem 287:14631–14643Google Scholar
- (9)Targeting the disordered C terminus of PTP1B with an allosteric inhibitorNat Chem Biol 10:558–566Google Scholar
- (10)Discovery of Small Molecules that Inhibit the Disordered Protein, p27Kip1Sci Rep 5:15686Google Scholar
- (11)EPI-001, A Compound Active against Castration-Resistant Prostate Cancer, Targets Transactivation Unit 5 of the Androgen ReceptorACS Chem Biol 11:2499–2505Google Scholar
- (12)Fasudil attenuates aggregation of α-synuclein in models of Parkinson’s diseaseActa Neuropathol Commun 4:39Google Scholar
- (13)Molecular Mechanism for the (−)-Epigallocatechin Gallate-Induced Toxic to Nontoxic Remodeling of Aβ OligomersJ Am Chem Soc 139:13720–13734Google Scholar
- (14)Elucidating the Aβ42 Anti-Aggregation Mechanism of Action of Tramiprosate in Alzheimer’s Disease: Integrating Molecular Analytical Methods, Pharmacokinetic and Clinical DataCNS Drugs 31:495–509Google Scholar
- (15)Identification of a Drug Targeting an Intrinsically Disordered Protein Involved in Pancreatic AdenocarcinomaSci Rep 7:39732Google Scholar
- (16)Principal component analysis of data from NMR titration experiment of uniformly (15)N labeled amyloid beta (1-42) peptide with osmolytes and phenolic compoundsArch Biochem Biophys 690:108446Google Scholar
- (17)Computational strategy for intrinsically disordered protein ligand design leads to the discovery of p53 transactivation domain I binding compounds that activate the p53 pathwayChem Sci 12:3004–3016Google Scholar
- (18)EGCG binds intrinsically disordered N-terminal domain of p53 and disrupts p53-MDM2 interactionNat Commun 12:986Google Scholar
- (19)A FRET-Based Biosensor for the Src N-Terminal Regulatory ElementBiosensors 12:96Google Scholar
- (20)Molecular Basis of Small-Molecule Binding to alpha-SynucleinJ Am Chem Soc 144:2501–2510Google Scholar
- (21)Tuning the rate of aggregation of hIAPP into amyloid using small-molecule modulators of assemblyNat Commun 13:1040Google Scholar
- (22)Rational optimization of a transcription factor activation domain inhibitorNat Struct Mol Biol 30:1958–1969Google Scholar
- (23)Picosecond Dynamics of a Small Molecule in Its Bound State with an Intrinsically Disordered ProteinJ Am Chem Soc 146:2319–2324Google Scholar
- (24)Structure-based Inhibitor Design for the Intrinsically Disordered Protein c-MycSci Rep 6:22298Google Scholar
- (25)Small-molecule sequestration of amyloid-β as a drug discovery strategy for Alzheimer’s diseaseSci Adv 6Google Scholar
- (26)Small Molecule Modulation of Intrinsically Disordered Proteins Using Molecular Dynamics SimulationsJ Chem Inf Model 60:5003–5010Google Scholar
- (27)Melatonin Inhibits hIAPP Oligomerization by Preventing β-Sheet and Hydrogen Bond Formation of the Amyloidogenic Region Revealed by Replica-Exchange Molecular Dynamics SimulationInt J Mol Sci 23Google Scholar
- (28)Small molecules targeting the disordered transactivation domain of the androgen receptor induce the formation of collapsed helical statesNat Commun 13:6390Google Scholar
- (29)Unveiling molecular mechanism underlying inhibition of human islet amyloid polypeptide fibrillation by benzene carboxylic acid-peptide conjugateJ Mol Liq 416:126426Google Scholar
- (30)A druggable conformational switch in the c-MYC transactivation domainNat Commun 15:1865Google Scholar
- (31)Predicting the sequence-dependent backbone dynamics of intrinsically disordered proteinseLife 12:RP88958https://doi.org/10.7554/eLife.88958Google Scholar
- (32)Atomistic molecular dynamics simulations of intrinsically disordered proteinsCurr Opin Struct Biol 92:103029Google Scholar
- (33)Sequence-Dependent Backbone Dynamics of Intrinsically Disordered ProteinsJ Chem Theory Comput 18:6310–6323Google Scholar
- (34)Long-Range Interactions Within a Nonnative ProteinScience 295:1719–1722Google Scholar
- (35)Ligand binding to distinct states diverts aggregation of an amyloid-forming proteinNat Chem Biol 7:730–739Google Scholar
- (36)Structure of alpha-helical membrane-bound human islet amyloid polypeptide and its implications for membrane-mediated misfoldingJ Biol Chem 283:17205–17210Google Scholar
- (37)Small-Molecule MYC Inhibitors Suppress Tumor Growth and Enhance ImmunotherapyCancer Cell 36:483–497Google Scholar
- (38)Synthetic fluorescent MYC probe: Inhibitor binding site elucidation and development of a high-throughput screening assayBioorg Med Chem 42:116246Google Scholar
- (39)MYC-Targeting Inhibitors Generated from a Stereodiversified Bicyclic Peptide LibraryJ Am Chem Soc 146:1356–1363Google Scholar
- (40)EGCG redirects amyloidogenic polypeptides into unstructured, off-pathway oligomersNat Struct Mol Biol 15:558–566Google Scholar
- (41)Ion Mobility Spectrometry–Mass Spectrometry Defines the Oligomeric Intermediates in Amylin Amyloid Formation and the Mode of Action of InhibitorsJ Am Chem Soc 136:660–670Google Scholar
- (42)The Flavanol (−)-Epigallocatechin 3-Gallate Inhibits Amyloid Formation by Islet Amyloid Polypeptide, Disaggregates Amyloid Fibrils, and Protects Cultured Cells against IAPP-Induced ToxicityBiochemistry 49:8127–8133Google Scholar
- (43)The green tea polyphenol (−)-epigallocatechin gallate prevents the aggregation of tau protein into toxic oligomers at substoichiometric ratiosFEBS Lett 589:77–83Google Scholar
- (44)Inhibition by Flavonoids of Amyloid-like Fibril Formation by Plasmodium falciparum Merozoite Surface Protein 2Biochemistry 49:5899–5908Google Scholar
- (45)Ensemble docking for intrinsically disordered proteinsbioRxiv https://doi.org/10.1101/2025.01.23.634614Google Scholar
- (46)Probabilistic sampling of protein conformations: new hope for brute force?Proteins 46:8–23Google Scholar
- (47)SwissDock 2024: major enhancements for small-molecule docking with Attracting Cavities and AutoDock VinaNucl Acids Res 52:W324–W332Google Scholar
- (48)AutoDock Vina 1.2.0: New Docking Methods, Expanded Force Field, and Python BindingsJ Chem Inf Model 61:3891–3898Google Scholar
Article and author information
Author information
Version history
- Sent for peer review:
- Preprint posted:
- Reviewed Preprint version 1:
Cite all versions
You can cite all versions using the DOI https://doi.org/10.7554/eLife.107470. This DOI represents all versions, and will always resolve to the latest one.
Copyright
© 2025, MacAinsh et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.
Metrics
- views
- 0
- downloads
- 0
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.