Fact-checked by Grok 2 weeks ago

Intrinsically disordered proteins

Intrinsically disordered proteins (IDPs), also known as natively unfolded or intrinsically unstructured proteins, are biologically active proteins or protein segments that lack a stable, fixed three-dimensional structure under physiological conditions, instead existing as dynamic ensembles of unfolded, collapsed, or extended non-globular conformations. These proteins are distinguished from traditional globular proteins by their high conformational flexibility and absence of persistent secondary or structures, enabling them to adopt diverse shapes in response to environmental cues or binding partners. A hallmark of IDPs is their biased amino acid composition, featuring enrichment in disorder-promoting residues such as , , , , , , , and serine, while being depleted in bulky hydrophobic like , , , , , , and . This composition results in low overall hydrophobicity, high net charge at physiological , and low sequence complexity, which collectively contribute to their structural and resistance to aggregation under normal conditions. IDPs often contain intrinsically disordered regions (IDRs), which can span entire proteins or form flexible linkers and tails within otherwise structured molecules, allowing for rapid adaptability in molecular interactions. IDPs play critical roles in numerous cellular processes, including molecular recognition, signaling, and , where their disorder facilitates promiscuous binding, entropic chain behavior as flexible linkers, and serving as sites for post-translational modifications. For instance, they function as assemblers in forming dynamic protein complexes, disordered chaperones to prevent aggregation, scavengers for neutralization, and even as metal sponges for storage. Their functional versatility often arises through coupled folding upon binding, where IDPs transiently adopt ordered structures only when interacting with partners, enhancing specificity and efficiency in pathways like transcription, , and control. In terms of prevalence, IDPs and IDRs are abundant across proteomes, constituting approximately 30–50% of eukaryotic proteins with long disordered regions exceeding 30 residues, with prevalence increasing alongside genome complexity—from about 2% in and 4% in eubacteria to higher levels in multicellular organisms. They are particularly enriched in the of cells, where they support macromolecular recognition and signaling, and are overrepresented in disease-associated proteins, such as those implicated in neurodegeneration and cancer, highlighting their biomedical significance.

Definition and Characteristics

Core Definition

Intrinsically disordered proteins (IDPs) are biologically active proteins or protein regions that lack a stable, fixed three-dimensional structure under physiological conditions of temperature, pH, and ionic strength. Instead, they exist as dynamic ensembles of interconverting conformations, including unfolded, partially collapsed, or extended states, which enable functional adaptability without relying on a rigid . In contrast to globular or ordered proteins, which fold into compact, stable structures driven by a hydrophobic core and specific intramolecular interactions essential for their enzymatic or binding functions, IDPs do not achieve such a defined architecture. This structural plasticity distinguishes IDPs, which comprise approximately 30–40% of residues in eukaryotic proteomes, often as long disordered segments exceeding 30 . The biophysical origins of intrinsic disorder stem from distinctive compositions: IDPs exhibit low sequence complexity, high net charge (positive or negative), and reduced overall hydrophobicity, leading to an absence of the hydrophobic core that stabilizes folded proteins. They are enriched in polar and charged residues such as , , , , serine, and , while depleted in bulky hydrophobic residues like , , , , and , which favor ordered secondary structures. Archetypal examples include the N-terminal of the tumor suppressor , which remains disordered to facilitate promiscuous interactions with multiple transcription factors, and the N-terminal projection of , a that adopts ensemble conformations critical for its regulatory roles.

Key Properties

Intrinsically disordered proteins (IDPs) exhibit distinctive sequence compositions that promote structural flexibility. These sequences are typically enriched in polar and charged , such as (), glycine (Gly), and serine (Ser), while depleted in bulky hydrophobic residues like tryptophan (Trp) and phenylalanine (Phe). This bias reduces the hydrophobic core formation essential for stable folding, favoring an extended, unstructured state. A key tool for classifying IDPs based on these sequence traits is the charge-hydropathy plot (CH-plot), which plots the mean net charge against the mean hydropathy of a protein sequence. In this plot, IDPs occupy regions of high net charge and low mean hydropathy, distinguishing them from folded proteins that cluster in areas of low charge and high hydropathy. This separation arises because the electrostatic repulsion from charged residues and the lack of hydrophobic driving forces prevent collapse into a compact . Biophysically, IDPs are characterized by high conformational due to their ability to sample a vast array of conformations without energetic barriers to transitions. This is reflected in rapid fluctuations on picosecond-to-nanosecond timescales, driven by local backbone and side-chain motions. The overall size of IDPs scales with chain length according to a R_g \sim N^{0.588}, indicative of denatured-like behavior with effects, in contrast to the R_g \sim N^{0.33} for compact, folded globular proteins. IDPs display sensitivity to environmental factors that modulate their degree of compaction without inducing folding. Changes in alter the protonation state of charged residues, influencing electrostatic interactions and thus chain extension or collapse. Similarly, increasing screens charges, promoting compaction in polyampholytic IDPs, while macromolecular crowders like induce effects that compact the chain through depletion attractions. These responses highlight how IDPs maintain disorder while adapting their ensembles to cellular conditions. Spectroscopically, IDPs show characteristic signatures consistent with their lack of ordered secondary . In (NMR) spectra, the absence of a fixed leads to narrow linewidths and limited dispersion due to fast tumbling and averaging over conformations. (CD) spectra of IDPs feature a strong negative band near 200 nm from disordered bonds but a weak or absent signal at 222 nm, indicating minimal α-helical content. (SAXS) profiles of IDPs exhibit a relatively shallow power-law at high scattering vectors q (exponent ≈ -2 to -3), reflecting smooth mass distributions without sharp solvent-exposed surfaces typical of folded proteins.

Historical Development

Early Discoveries

In , initial observations of protein denaturation challenged the emerging lock-and-key model of action by showing that unfolded proteins could sometimes retain partial or regain it upon reversal of denaturing conditions. Researchers, including Alfred E. Mirsky and , demonstrated the reversibility of denaturation in proteins like and , where removal of heat or allowed reformation of native properties, suggesting the unfolded state was a dynamic ensemble rather than a permanent loss of function. These findings, emerging from biophysical studies at the Rockefeller Institute for Medical Research and the Gates Chemical Laboratory at the , laid groundwork for recognizing flexible protein states but were largely interpreted within the framework of transient unfolding rather than inherent disorder. The 1950s and 1960s brought more systematic evidence through enzymatic studies and spectroscopic methods. Christian Anfinsen's experiments on bovine pancreatic ribonuclease at the National Institutes of Health revealed that fully denatured and reduced ribonuclease, stripped of its disulfide bonds, could spontaneously refold and regain full enzymatic activity in vitro, underscoring the amino acid sequence as the sole determinant of structure. Although focused on refolding pathways, this work implied functional relevance for unfolded intermediates. Concurrently, the advent of nuclear magnetic resonance (NMR) spectroscopy provided early glimpses of protein dynamics in solution; the first NMR spectrum of a protein (ribonuclease A) was reported in 1957, and subsequent 1960s studies detected motional flexibility in side chains and backbones that contrasted with the static views from X-ray crystallography. By the 1970s and 1980s, techniques such as and spectroscopy identified proteins existing in "natively unfolded" or partially structured states under physiological conditions. For example, prothrombin fragment 1, a key component in blood coagulation, was shown to lack significant ordered secondary structure in the absence of calcium ions, adopting a flexible, extended conformation that enables binding upon ion coordination. The "molten globule" concept, proposed by Oleg B. Ptitsyn, described compact folding intermediates with native-like secondary structure but fluctuating tertiary contacts, observed in denaturation/renaturation experiments on globular proteins like alpha-lactalbumin. Jane S. Richardson's 1981 analysis of known protein structures via ribbon diagrams explicitly noted extensive flexible loops and irregular regions comprising up to 40% of polypeptide chains, yet these observations faced resistance due to the entrenched dogma that functional proteins require unique, stable folds.

Modern Recognition

In the 1990s, advances in (NMR) spectroscopy and began to challenge the traditional view that all functional proteins require stable three-dimensional structures, leading to the recognition of a class of proteins lacking fixed folds under physiological conditions. Peter E. Wright and H. Jane Dyson introduced the term "intrinsically unstructured proteins" in their seminal 1999 review, arguing that such proteins represent a distinct functional category rather than denatured or anomalous states, and calling for a reassessment of the protein structure-function paradigm. Concurrently, Vladimir N. Uversky and colleagues published key studies on natively unfolded proteins, such as the , highlighting their roles in diseases and functional plasticity. Computational tools emerged to predict disorder from sequences; the Predictor of Natural Disordered Regions (PONDR), developed by Pedro Romero and colleagues in 1997, used neural networks to identify disordered segments based on composition and complexity, enabling large-scale screening of proteomes. The early 2000s formalized the link between disorder and biological function, shifting perceptions from viewing these proteins as exceptions to essential components of cellular regulation. In 2001, A. Keith Dunker and Zoran Obradović coined the term "intrinsically disordered proteins" (IDPs) in a commentary proposing the "protein trinity" model—encompassing ordered, molten globule, and disordered states—and highlighting how disorder facilitates multifunctionality in signaling and regulatory roles. This conceptual framework was supported by experimental evidence showing that disorder enables adaptive binding and allosteric regulation. To catalog such proteins, the Database of Disordered Protein (DisProt) was established in 2005 by Srdan Vucetic and colleagues, providing a manually curated repository of IDP structural and functional annotations derived from literature. By the 2010s, IDPs were integrated into broader initiatives, reflecting their acceptance as a core protein class. Structural genomics projects, such as the Protein Structure Initiative, increasingly incorporated disorder predictions to prioritize targets and interpret NMR and data, as disorder often hindered but was crucial for ; a 2012 analysis demonstrated how intrinsic disorder knowledge improved target selection and structure determination success rates in proteomics pipelines. This era also saw indirect validation through the 2013 awarded to , , and for multiscale computational modeling of complex systems, which advanced simulations of protein dynamics essential for understanding IDP ensembles. Recent updates in the 2020s have further solidified this recognition, with analyses indicating that approximately 50% of regulatory and signaling proteins contain significant disordered regions, underscoring their prevalence in eukaryotic control networks. For instance, as of 2025, advances in and simulations have enabled accurate ensemble predictions and de novo design of IDPs with tailored properties, enhancing therapeutic applications. This progression marked a from dismissing disordered proteins as structural anomalies to embracing them as functionally vital, with influential reviews reinforcing the change; for instance, a 2014 overview by Christopher J. Oldfield and A. Keith Dunker in Annual Review of Biochemistry synthesized evidence that IDPs drive adaptive cellular responses through conformational plasticity.

Prevalence in Nature

Abundance Across Organisms

Intrinsically disordered proteins (IDPs) and regions (IDRs) exhibit varying prevalence across the domains of life, with eukaryotes displaying significantly higher levels than prokaryotes. In , approximately 2% of proteins contain long disordered regions exceeding 30 residues, while in this figure rises to about 4.2%; in contrast, eukaryotes harbor around 33% of such proteins. The fraction of disordered residues within proteomes further underscores this gradient, comprising roughly 30% in eukaryotes compared to less than 27% in most archaeal and bacterial species. These patterns reflect a broader trend where full-length IDPs—defined as proteins with over 50% disordered residues—are estimated at 6-8% in select archaeal genomes, 4.5-5% in bacterial ones, and 23-28% in eukaryotic examples like and mammals. Within eukaryotes, the abundance of disorder escalates in more complex organisms, particularly metazoans. For instance, human proteomes feature disordered residues in approximately 45% of amino acids, with signaling proteins showing even higher enrichment—66% containing long IDRs of at least 30 residues. This elevated disorder supports dynamic interactions in multicellular signaling networks. Comprehensive datasets like DisProt (version 9, 2023, with 2,649 curated protein entries; as of version 9.8 in June 2025, 3,201 entries) and (updated September 2025 with 232 new entries) document these patterns, aggregating experimental evidence from techniques such as NMR and to annotate IDPs across species. IDRs are notably enriched in specific functional domains, particularly nuclear proteins and transcription factors, where disorder facilitates promiscuous binding to DNA and regulatory partners. Analysis of DNA- and RNA-binding proteins across 1,121 species reveals that these nuclear-associated factors are significantly more disordered than non-binding counterparts, with eukaryotes showing the highest levels. Conversely, IDPs are scarce in membrane proteins, which typically require ordered transmembrane helices for lipid embedding and stability; disorder content in integral membrane proteomes is thus markedly lower, often confined to flexible intracellular loops rather than full proteins. This underrepresentation aligns with the biophysical constraints of membrane environments, limiting widespread disorder in such contexts.

Evolutionary Perspectives

Intrinsically disordered proteins (IDPs) and their regions (IDRs) exhibit a marked increase in prevalence across phylogenetic lineages, rising from approximately 8.5% of the in prokaryotes to 20.5% in eukaryotes. This gradient reflects the growing complexity of cellular regulation in more advanced organisms, where IDPs contribute to proteome expansion through mechanisms such as and insertions/deletions that preferentially incorporate disordered segments. Additionally, plays a pivotal role in generating diverse disordered isoforms, thereby enhancing functional versatility without requiring extensive sequence innovation. Despite their overall rapid evolutionary rates, IDR regions demonstrate selective conservation patterns that distinguish them from structured domains. IDRs evolve faster than ordered protein domains, characterized by higher rates and distinct residue preferences enriched in serine and proline, yet they maintain constraints on distributed physicochemical features such as flexibility and charge patterns. Functional motifs within IDRs, including short linear motifs (SLiMs), show evidence of preservation, with over 25% of disordered sites evolving more slowly than comparable ordered sites due to their roles in molecular . This higher degree of in eukaryotes correlates strongly with the of multicellularity, enabling intricate regulatory architectures that demand adaptive flexibility. The evolutionary persistence of IDPs confers selective advantages by facilitating the rapid rewiring of regulatory networks, allowing organisms to adapt swiftly to environmental pressures through point mutations and indels that alter profiles without disrupting functions. A prominent example is the tails, which are intrinsically disordered and exhibit significant sequence divergence across species while retaining essential roles in and post-translational modifications. This divergence enables functional promiscuity and dosage sensitivity, promoting retention after duplication and supporting the of complex signaling pathways.

Structural Properties

Conformational Ensembles

Intrinsically disordered proteins (IDPs) exist as heterogeneous populations of rapidly interconverting conformers in solution, lacking a dominant folded and instead sampling a diverse array of conformations governed by the . The relative populations of these conformers are determined by their free energies, with average properties such as the reflecting the ensemble rather than any single state. This statistical description emphasizes that IDP behavior arises from ensemble-averaged observables, as no unique three-dimensional predominates. To model the conformational sampling of IDPs, polymer physics approaches such as the (WLC) model are employed, treating the polypeptide as a semi-flexible chain to predict properties like the , which quantifies the spatial extent of the ensemble. In the WLC framework, the persistence length—a measure of chain stiffness—varies with sequence composition, enabling predictions of how content or glycine-rich regions influence overall elasticity and compactness. For instance, sequences with high polyproline II propensity exhibit greater rigidity, resulting in larger compared to more flexible glycine-dominated chains. In crowded cellular environments, IDP ensembles undergo compaction primarily through excluded volume effects, where macromolecular crowders reduce the accessible space and favor more collapsed conformations. This entropic driving force can reduce the end-to-end distance or by up to 40% for moderately hydrophobic IDPs at crowder volume fractions around 40%, though the magnitude depends on sequence hydrophobicity and crowder properties. Such effects highlight how environmental factors modulate the intrinsic ensemble without inducing folding. Recent advances as of 2025, including improved force fields and AI-based methods like IDPFold, have enhanced the accuracy of generating and predicting IDP conformational ensembles from sequences, providing deeper insights into their structural diversity. A representative example is the , where the conformational ensemble spans extended coil-like states with minimal long-range contacts to more compact forms stabilized by transient electrostatic interactions, as captured by mean radii of gyration around 5.1 nm in isolation. In hTau40, the full-length isoform, NMR data reveal elevated polyproline II sampling in aggregation-prone regions alongside helical propensities in distant segments, contributing to a dynamic range from expanded to collapsed states. NMR-derived ensembles of IDPs, such as the CytR N-terminal , further illustrate millisecond-scale , with excited folded states populating about 14% of the ensemble and exchanging at rates of approximately 49 s⁻¹. These timescales reflect the rapid interconversion central to IDP ensembles.

Dynamic Features

Intrinsically disordered proteins (IDPs) exhibit a wide range of dynamic timescales that underpin their functional flexibility, spanning from sub-nanosecond local fluctuations in backbone and side-chain motions to microsecond-scale global rearrangements of chain segments. These rapid local dynamics, often probed by (NMR) relaxation techniques, reflect the high conformational characteristic of disordered states, allowing IDPs to sample diverse local geometries without stable secondary structures. Slower processes, such as segmental reorientations or loop formations, occur on timescales and are influenced by sequence-specific interactions like hydrophobic clustering or electrostatic repulsion. The temperature dependence of these dynamics typically follows Arrhenius-like behavior, with activation energies varying by residue type; for instance, polar residues show lower barriers (around 10-15 kJ/mol) compared to hydrophobic ones, enabling adaptive responses to environmental changes. Thermodynamically, IDPs are characterized by flat energy landscapes featuring multiple shallow minima and low barriers to interconversion, contrasting with the funnel-shaped landscapes of folded proteins. This rugged yet accessible terrain results in , where conformational populations are broadly distributed rather than dominated by a single state, as evidenced by ensemble averaging of dynamics across equilibrium conformations. () reveals distinctive anomalies in IDPs, such as broad, low-amplitude transitions or baseline elevations without sharp unfolding peaks, reflecting the absence of folding events and the exposure of hydrophobic groups to across temperatures. These features highlight how disorder minimizes energetic penalties for structural excursions, facilitating rapid . The intrinsic dynamics of IDPs enable pronounced responsiveness, including allosteric effects mediated without rigid structural domains, where perturbations at one site propagate through the flexible chain to influence distant regions. A hallmark is the disorder-to-order transition upon ligand binding, as seen in molecular recognition features (MoRFs) within IDPs like those in the , where binding induces partial folding on timescales of microseconds to milliseconds, enhancing specificity and . This entropic-driven allostery leverages the flat to couple local binding events to global conformational shifts, optimizing in pathways such as regulation. Such mechanisms underscore how IDP dynamics integrate environmental cues efficiently, distinct from the steric hindrance in ordered proteins.

Biological Functions

Flexible Linkers and Motifs

In multi-domain proteins, (IDPs) often function as flexible linkers that connect structured domains, enabling relative movement and modulating inter-domain orientations through their inherent conformational flexibility. These linkers act primarily as entropic chains, where their disordered nature provides an entropic penalty against extension, behaving like springs that resist deformation without requiring specific secondary structures. This flexibility allows the protein to adopt a wide range of conformations, facilitating adaptive responses to mechanical or environmental cues while maintaining overall stability. A prominent example of such entropic springs is found in the giant muscle protein , where the PEVK-rich region—composed of , glutamate, , and residues—serves as a disordered linker between immunoglobulin-like domains in the I-band. Upon muscle stretching, this region extends, generating restoring force through entropic elasticity to aid in recoil and prevent overstretching. Similarly, in the complex, FG-nucleoporins (FG-Nups) feature long intrinsically disordered regions with phenylalanine-glycine repeats that act as flexible linkers, anchoring structured cores while extending into the central channel to form a dynamic barrier for selective nucleocytoplasmic transport. These linkers' disordered state enables transient interactions and rapid reconfiguration, essential for the pore's high-throughput function. Beyond extended linkers, IDPs frequently contain short linear motifs (SLiMs), which are brief disordered sequences of 3–10 residues that mediate low-affinity, specific interactions with partner proteins. These motifs are embedded within flexible regions, allowing accessibility without the energetic cost of stable folding, and are particularly prevalent in signaling pathways. A classic instance is the PxxP motif recognized by SH3 domains, where the core proline-rich sequence (e.g., in class I: R/KxxPxxP) binds to adaptors like or , facilitating transient assembly in cascades such as MAPK signaling; such motifs show enhanced conservation in disordered contexts compared to ordered ones, underscoring their functional importance. The functional diversity of these elements in IDPs arises from their dual roles: as spacers that promote efficient macromolecular by preventing steric clashes and maintaining optimal spacing, and as motifs that confer specificity at minimal energetic expense, enabling rapid on-off ideal for regulatory networks. In contexts, linkers like those in cytoskeletal proteins ensure proper geometric arrangement, while SLiMs provide modular docking sites that evolve quickly yet remain functionally robust. This versatility highlights how disorder supports both structural adaptability and precise molecular recognition in cellular architecture.

Coupled Binding Mechanisms

Intrinsically disordered proteins (IDPs) often engage in coupled folding and processes, where they transition from a disordered state to an ordered conformation upon interacting with a binding partner. This disorder-to-order transition mitigates the entropic penalty associated with folding by coupling it directly to the enthalpically favorable event, allowing IDPs to achieve high-affinity interactions without the need for prior independent folding. The fly-casting mechanism exemplifies this kinetic advantage: the extended, disordered chain increases the effective capture radius for the target, enabling rapid initial contacts that guide subsequent folding and tight , as demonstrated in theoretical models of unstructured protein association. Pre-structured motifs within IDPs further facilitate efficient binding by adopting transient secondary structures, such as helices or turns, even in the unbound state, which prime the protein for recognition. These motifs lower the activation barrier for folding upon binding by pre-organizing key interaction sites. A prominent example is the (TAD) of , which exhibits a weak propensity for helical structure in residues 18–26 in its free form, enabling rapid helical folding when binding to the N-terminal domain of ; this pre-structuring enhances specificity and affinity in the p53-MDM2 regulatory complex. In contrast to fully ordered complexes, fuzzy complexes represent a mode where IDPs retain significant disorder even after binding, featuring dynamic interfaces that allow structural heterogeneity and functional versatility. These ensembles of conformations enable adaptive interactions, often with multiple binding modes coexisting. For instance, the interaction between the intrinsically disordered inhibitor IκBα and the NF-κB p50/RelA heterodimer forms a fuzzy complex, where IκBα's ankyrin repeats maintain flexibility while masking the nuclear localization signals of p50 and RelA, facilitating regulated nuclear transport and transcriptional control.

Roles in Phase Separation

Intrinsically disordered proteins (IDPs) play a central role in liquid-liquid (LLPS), a process that drives the formation of biomolecular condensates, which are membraneless intracellular compartments. These condensates enable dynamic organization of cellular processes without the need for lipid membranes. The mechanism of LLPS in IDPs relies on multivalent weak interactions mediated by intrinsically disordered regions (IDRs), where specific residues act as "stickers" that form transient contacts, interspersed with flexible "spacers" that modulate chain dynamics and solubility. This stickers-and-spacers framework explains how low-affinity, multivalent interactions between IDR segments promote the coalescence of proteins into liquid-like droplets, as demonstrated in biophysical models of IDP . A prominent example of IDPs driving LLPS is the RNA-binding proteins FUS and TDP-43, which contain prion-like domains rich in stickers such as arginine-glycine-glycine (RGG) motifs and residues. These domains facilitate phase separation into stress granules, dynamic condensates that sequester mRNAs and translation factors during cellular stress, thereby regulating . The multivalency arises from the flexible linkers and motifs in these IDRs, allowing rapid association and disassembly of droplets to respond to environmental cues. In biological contexts, IDP-driven condensates serve essential functions in compartmentalization and signaling. For instance, the , a key site of , forms through LLPS of IDPs like nucleophosmin, concentrating ribosomal proteins and RNAs to enhance assembly efficiency. Similarly, , involved in mRNA decay and storage, rely on IDPs such as tristetraprolin to sequester transcripts and regulatory factors, preventing unwanted . These condensates also regulate transcription by concentrating enhancers and transcription factors at loci, as seen in super-enhancer hubs, and modulate signaling pathways by dynamically partitioning kinases and substrates to control reaction rates. Recent advances from 2023 to 2025 have elucidated specific IDR sequence features governing phase behavior, including π-cation interactions between aromatic residues (e.g., , ) and positively charged like , which enhance multivalency and lower the critical concentration for LLPS. These interactions contribute to the of condensates while maintaining , as quantified in peptide-based studies showing up to 80% stabilization in high-aromatic, low-charge systems. Such insights have revealed how aberrant LLPS of IDPs like FUS and TDP-43 can lead to pathological condensates in neurodegeneration, highlighting the fine balance between physiological and dysfunctional . In 2025, protein language models have been employed to identify motifs within IDRs that act as evolutionary units supporting and formation.

Experimental Characterization

In Vitro Methods

In vitro methods provide controlled environments to characterize the structural and dynamic properties of intrinsically disordered proteins (IDPs) using purified samples, enabling precise measurements of conformational ensembles and flexibility without cellular interference. These techniques are essential for validating the absence of stable secondary or structures and quantifying heterogeneity in IDP populations. Spectroscopic approaches are widely employed to probe local and global features of IDPs. (CD) detects the lack of ordered secondary structure in IDPs by revealing minimal signal in the far-UV region, typically showing signatures rather than alpha-helical or beta-sheet patterns. For instance, CD spectra of IDPs like exhibit low ellipticity around 200 nm, confirming their disordered state under physiological conditions. , particularly Förster resonance energy transfer (FRET), assesses compaction and intramolecular distances in IDPs; single-molecule FRET reveals heterogeneous conformations and transient compact states in proteins such as . (NMR) offers atomic-level insights into IDP ensembles through deviations and relaxation rates; for example, ¹H-¹⁵N HSQC spectra of IDPs display narrow dispersion indicative of disorder, while R₁ and R₂ relaxation measurements quantify ps-ns timescale dynamics. These methods collectively demonstrate that IDPs populate diverse conformers rather than rigid folds. Scattering and hydrodynamic techniques evaluate the overall dimensions and solvent interactions of IDPs in solution. (SAXS) determines (R_g) and maximum dimension (D_max), revealing expanded conformations in IDPs; for example, SAXS profiles of disordered domains show power-law decays consistent with flexible chains, with R_g values often 1.5–2 times larger than globular proteins of similar mass. (SANS) complements SAXS by probing solvent effects through contrast variation, highlighting hydration layers around IDPs like disordered tails in proteins. (AUC), particularly sedimentation velocity, measures hydrodynamic radii and molecular weights, confirming extended shapes in IDPs; sedimentation coefficients (s) for proteins such as the intrinsically disordered reveal non-globular behavior with s-values lower than expected for compact forms. Despite their precision, methods have limitations, including artifacts from low concentrations that may stabilize non-native states or promote unwanted aggregation, and the absence of cellular crowding or binding partners that influence behavior . For , a prototypical linked to , aggregation assays like Thioflavin T fluorescence monitoring often suffer from poor reproducibility due to heterogeneous and sensitivity to trace impurities, potentially overestimating fibril formation rates outside physiological contexts.

In Vivo Techniques

In vivo techniques for studying intrinsically disordered proteins (IDPs) provide critical insights into their behavior within the complex cellular environment, where and dynamic interactions influence conformational ensembles and functions that differ from those observed . These methods emphasize live-cell or organismal contexts to validate disorder, mobility, and interactions, often integrating fluorescence-based imaging and perturbation strategies to capture transient states. Fluorescence recovery after photobleaching (FRAP) is widely used to assess IDP mobility and in living cells, particularly in phase-separated condensates or membrane-less organelles. For instance, FRAP measurements of the low-complexity domain of FUS revealed rapid exchange dynamics indicative of liquid-like behavior in stress granules, with recovery times on the order of seconds highlighting the role of disorder in facilitating material exchange. techniques, such as depletion (STED) or photoactivated localization microscopy (), enable visualization of IDP dynamics at nanoscale resolutions beyond limits, revealing spatial organization and transient clustering. In nucleocytoplasmic studies, high-speed of FG-nucleoporins—IDP-rich components of complexes—demonstrated bidirectional shuttling kinetics with residence times of milliseconds, underscoring disorder-driven selectivity. Förster resonance energy transfer (FRET), especially (smFRET) in cellular contexts, probes conformational changes and intramolecular distances in IDPs. Live-cell smFRET on α-synuclein showed population shifts toward more compact states under cellular crowding compared to dilute solutions, with transfer efficiencies indicating reductions of up to 20%. Perturbation approaches complement imaging by testing the functional necessity of disorder; can rigidify IDRs by introducing residues or stabilizing secondary structures, allowing assessment of impacts on cellular processes. For example, mutating the disordered activation domain of the Gcn4 in reduced target activation by over 50%, confirming the entropic role of flexibility in promoter binding. Chemical (XL-MS) maps transient interactions of by capturing proximity under native conditions. Membrane-permeable cross-linkers like disuccinimidyl suberate (DSS) enable proteome-wide profiling in intact cells, identifying partners such as α-synuclein's interactions with synaptic proteins in neuronal cells, with cross-link yields revealing dynamic interfaces not detectable by purification. Despite these advances, challenges persist due to , which occupies 20-30% of cellular volume and can compact ensembles, altering hydrodynamic radii by factors of 1.5-2 as seen in in-cell NMR studies. Transient states further complicate detection, as sub-millisecond fluctuations evade conventional averaging. Emerging live-cell NMR techniques address these issues by providing atomic-resolution dynamics directly , particularly for . In human cells, in-cell NMR of full-length tau revealed phosphorylation-induced shifts in residue-specific chemical shifts, indicating enhanced disorder in the microtubule-binding repeats with linewidth broadenings reflecting crowding effects; these 2020s developments, building on models, now enable monitoring of tau's conformational plasticity in neurodegeneration-relevant contexts.

Computational Modeling and Prediction

Molecular Simulations

Molecular dynamics (MD) simulations have become essential for modeling the conformational dynamics and structural ensembles of intrinsically disordered proteins (IDPs), capturing their flexibility at atomic resolution over timescales inaccessible to experiments. These simulations rely on empirical force fields to define interatomic interactions, with all-atom approaches providing detailed insights into sequence-specific behaviors. A prominent all-atom for IDP simulations is AMBER ff14SB, which balances backbone and side-chain parameters to reproduce disordered conformations without excessive compaction. Refinements like ff14IDPs further optimize potentials for better sampling of extended states, enabling simulations of IDPs such as α-synuclein to match experimental radii of gyration. For longer timescales, coarse-grained models reduce complexity by representing residues as beads, with the AWSEM-IDP incorporating knowledge-based potentials tuned for IDP hydrophobicity and charge patterns to simulate multiscale dynamics efficiently. Enhanced sampling techniques address the rugged energy landscapes of IDPs; replica-exchange MD (REMD) swaps configurations between temperature replicas to overcome kinetic barriers, generating diverse ensembles for proteins like the N-terminal domain of the . These methods facilitate computation of landscapes, revealing minima corresponding to transient helices or compact states in IDPs. Simulated ensembles are validated against experimental data from NMR and (SAXS), such as chemical shifts and scattering profiles, to ensure physical realism—often using to reweight trajectories for agreement with measurements. For instance, REMD simulations of the disordered protein have reproduced SAXS curves and NMR relaxation rates, confirming the heterogeneity of its conformational distribution. Recent advances (2023–2025) have integrated MD with multiscale modeling to probe IDP-driven phase separation, incorporating electrostatics and hydrophobic interactions to simulate condensate formation in proteins like FUS. Coarse-grained enhancements, such as those in AWSEM, now capture sequence-dependent liquid-liquid phase transitions over microseconds, validated against fluorescence recovery after photobleaching data. These developments provide atomistic views of multivalent interactions underlying biomolecular condensates.

Sequence-Based Prediction

Sequence-based prediction methods identify intrinsically disordered protein (IDP) regions solely from primary sequences, relying on patterns in composition, physicochemical properties, or learned features without requiring structural or evolutionary data. These approaches emerged in the early to address the challenge of detecting in proteins lacking stable folds, which traditional structure tools often overlook. Compositional methods, for instance, exploit biases such as low sequence complexity or enrichment in charged/hydrophobic residues, as disordered regions typically show reduced hydrophobicity and higher net charge compared to globular domains. Tools like low-complexity scorers (e.g., based on repeat detection) flag potential by quantifying repetitive or biased motifs that hinder stable packing. Machine learning-based predictors build on these features by training models to estimate propensity. A prominent example is IUPred, which uses an to score inter-residue interactions, assuming disordered regions exhibit weak pairwise energies similar to unfolded states rather than the stabilizing contacts in ordered structures. This method employs a support vector machine-like approach on estimated interaction energies derived from windows, achieving robust predictions for long disordered segments. Meta-predictors like PONDR (Predictor of Naturally Disordered Regions) integrate multiple algorithms, such as neural networks trained on compositional and local features, to enhance accuracy by averaging outputs from diverse models like VSL2 or VL-XT. Another example, DisEMBL, distinguishes types (e.g., regions versus molten globules) using neural networks on profiles, particularly effective for differentiating globular from flexible loop-like . Performance of these methods typically yields area under the curve () values around 0.8 on datasets, indicating good but imperfect discrimination between ordered and disordered residues; for instance, IUPred short and long variants achieve AUCs of 0.79 and 0.79, respectively, while PONDR VSL2B reaches 0.90 on certain sets. However, limitations arise with short disordered motifs (under 30 residues), where false negatives are common due to insufficient sequence context for reliable scoring, and predictions often over- or under-estimate boundary regions. DisEMBL, for example, excels in identifying flexible loops ( ~0.72) but struggles with binding-induced disorder. Validation involves ing against curated databases: structured regions from the (PDB) serve as negative controls, while DisProt provides experimentally verified disordered annotations from techniques like NMR and CD . These predictors generally do not account for post-translational modifications (PTMs) like , which can induce or stabilize disorder, leading to discrepancies where PTMs alter conformational ensembles.

AI and Machine Learning Advances

While has transformed , it struggles with intrinsically disordered proteins (IDPs) primarily due to its design for outputting single, rigid conformations, which fails to capture the dynamic ensembles characteristic of IDPs, compounded by difficulties in multiple sequence alignments from their hypervariable sequences. To overcome this, the -Metainference method, developed in 2025, employs 's predicted inter-residue distances as Bayesian restraints in metainference simulations, generating accurate conformational ensembles for IDPs that match experimental observables like (SAXS) and (NMR) data. This extension leverages 's strength in distance prediction despite disorder, enabling high-resolution modeling of IDP structural landscapes without relying solely on physics-based sampling. Machine learning has also advanced IDP function annotation, with the 2023 DisProt database release expanding functional coverage through enhanced curation and integration of ML-driven predictors like IDP-LM, a protein that forecasts disorder and associated functions such as or from sequences alone, achieving improved accuracy over traditional methods. In experimental realms, facilitates NMR for IDPs; for instance, the 2025 HyperW-Decon algorithm processes hyperpolarized water NMR spectra using convolutional neural networks to resolve transient ion interactions in disordered proteins involved in , preserving while accessing short-lived intermediates previously undetectable due to sensitivity limits. These tools enhance the interpretability of IDP dynamics in solution. Generative AI models have revolutionized IDP design, particularly through diffusion-based approaches that create novel binders for disordered targets. A 2025 study in Nature introduced a sequence-conditioned diffusion model, RFdiffusion-IDP, which generates de novo protein binders to IDPs like amylin without predefined scaffolds, yielding structures with sub-nanomolar affinities confirmed by calorimetry and crystallography, thus enabling targeting of previously "undruggable" flexible regions. Complementing this, a Harvard-Northwestern collaborative method from 2025 uses generative machine learning with automatic differentiation on coarse-grained polymer physics to inversely design intrinsically disordered regions (IDRs) with prescribed ensemble properties, such as compactness or responsiveness to stimuli, producing sequences that match target distributions in simulations and experiments for applications in sensors and linkers. These innovations bridge sequence-to-function gaps, prioritizing data-driven generation over exhaustive enumeration.

Annotation and Databases

Disorder Databases

DisProt serves as the central manually curated repository for intrinsically disordered proteins (IDPs) and regions (IDRs), compiling experimental evidence on their structural and functional properties from the literature. Launched in 2011, the database emphasizes high-quality annotations supported by standardized evidence, with version 9.8 (released in June 2025) containing 3,201 protein entries and over 11,000 pieces of evidence for disorder, reflecting continued growth since 2023. Annotations detail disordered regions with specific evidence codes, such as (NMR) spectroscopy for conformational dynamics and (CD) for secondary structure absence, ensuring traceability to original experiments. Entries are cross-linked to for sequence data and the (PDB) for associated structured domains, facilitating integrated analysis of full-length proteins. Curation in DisProt adheres to the Minimum Information About Disorder Experiments (MIADE) guidelines, which standardize reporting of experimental conditions, methods, and states to enhance and across resources. Between 2023 and 2025, DisProt expanded with thematic datasets and increased annotations linking to contexts, supported by a growing community. The database complements DisProt by focusing on structural ensembles of IDPs and IDRs, providing manually curated data on experimentally verified , binding partners, and post-translational modifications. It aggregates knowledge from biophysical studies, emphasizing dynamic conformations rather than static structures, with annotations cross-referenced to and external databases like . Updated to version 08/Sep/2025, maintains a collection of 1,342 entries, prioritizing quality through evidence-based validation from techniques like NMR and . MobiDB integrates curated disorder annotations from sources like DisProt and with computational predictions across the entire (over 227 million sequences as of 2025), offering a comprehensive view of intrinsic . Its annotations include experimentally derived regions with evidence from missing in PDB structures or direct biophysical assays (e.g., NMR, ), alongside predicted disorder scores, and provide direct cross-links to accessions and PDB files for contextual exploration. The 2023 update marked a decade of development, enhancing integration of ensemble properties and expanding PDB-derived annotations to 65,290 proteins as of 2025, while maintaining synchronization with for broad accessibility. These databases collectively enable the validation of sequence-based predictions by providing gold-standard experimental benchmarks.

Functional Annotation

Functional annotation of intrinsically disordered proteins (IDPs) and regions (IDRs) primarily relies on curated databases that extend structural evidence with functional descriptors, such as short linear motifs (SLiMs), interaction partners, and regulatory mechanisms. SLiM-based annotations identify short, sequence-specific motifs within disordered regions that mediate interactions with structured domains or other partners, often integrated from the Eukaryotic Linear (ELM) resource, which catalogs 356 motif classes enriched in IDRs (as of 2024). These motifs facilitate functions like signaling and trafficking, with ELM providing experimental validation for motif instances in eukaryotic proteomes. Binding partner annotations link IDRs to specific interactors, such as in DisProt, where regions are tagged with known protein, , or partners derived from curation. Regulatory roles are annotated through terms describing entropic chain functions, like chaperone activity or propensity, emphasizing the non-specific contributions of to cellular processes. The DisProt database has expanded its functional ontology in recent years, incorporating the Intrinsically Disordered Proteins Ontology (IDPO) alongside (GO) terms to standardize annotations for over 3,900 disordered regions across 1,300 proteins as of 2024, enabling of functions like molecular recognition and assembly. This expansion includes 172 new GO annotations and 63 IDPO terms in its 2025 release, focusing on context-specific roles while building on core disorder evidence from integrated structural databases. Annotating IDR functions presents challenges due to their context-dependency, where the same disordered segment can adopt varied conformations and roles based on cellular environment, partners, or post-translational modifications, complicating standardized curation. Multifunctionality further hinders annotation, as IDRs often bind multiple partners promiscuously, performing diverse tasks like signaling and within a single protein, which requires nuanced, evidence-based tagging to avoid oversimplification. Tools like address these by predicting disordered binding regions (DBRs) that indicate potential motif-domain sites, using energy-based scoring to highlight segments prone to induced folding upon , thus aiding in the identification of functional hotspots. Advances in (ML) have accelerated functional of uncharacterized IDRs, particularly in , with methods leveraging protein language models to predict propensities and conserved motifs directly from sequences. For instance, MobiDB's update integrates ML-driven ensemble predictions with curated functions, employing three strategies—GO mapping, SLiM detection, and forecasting—to annotate disordered content in eukaryotic proteomes, prioritizing high-confidence regulatory roles. Sequence-based ML tools, such as those using for intermolecular , further enable proteome-wide by scoring IDR-driven bindings, reducing reliance on experimental data for emerging proteomes.

Implications for Disease and Therapy

Disease Associations

Intrinsically disordered proteins (IDPs) and their regions (IDRs) are frequently implicated in human diseases through mechanisms involving gain-of-function via aberrant interactions or loss-of-function due to mutations that disrupt normal dynamics. In gain-of-function pathologies, IDRs can promote pathological aggregation, such as the formation of from the disordered N-terminal region of α-synuclein in , where misfolded aggregates lead to neuronal toxicity. Similarly, in and other neurodegenerative conditions, IDPs like and amyloid-β contribute to formation, exacerbating protein misfolding and cellular dysfunction. Loss-of-function often arises from mutations in IDRs that impair regulatory interactions; for instance, mutations in , including those in its intrinsically disordered that impair partner binding and transcriptional activation, contribute to cancer progression; is mutated in over 50% of human tumors overall. A high prevalence of disorder is observed among disease-associated proteins, with approximately 79% of cancer-related proteins and 66% of proteins containing long predicted disordered regions of 30 or more residues, far exceeding levels in the general . In neurodegeneration, IDPs are enriched, with many linked to amyloidogenic processes; for example, tau's disordered state enables hyperphosphorylation and detachment from in , leading to neurofibrillary tangles. Polyglutamine expansions, as in , alter the dynamics of the largely disordered protein, recruiting other ID domain-containing proteins into toxic aggregates and disrupting . These expansions, typically exceeding 35 glutamines, confer a dominant gain-of-function by enhancing aggregation propensity through intrinsically disordered domains. Recent studies highlight the role of IDP-driven biomolecular condensates in diseases like (ALS) and (FTD), where mutations in disordered low-complexity regions of proteins such as TDP-43 and FUS promote aberrant liquid-liquid , transitioning from functional droplets to pathological solid aggregates that impair processing and neuronal function. In ALS/FTD, these condensates sequester essential factors, contributing to neurodegeneration; for instance, TDP-43 mutations enhance formation within stress granules. Such misregulation underscores how IDP plasticity, when perturbed, underlies a spectrum of pathologies from cancer to inherited neurodegenerative disorders.

Therapeutic Targeting

Targeting intrinsically disordered proteins (IDPs) for therapeutic purposes presents significant challenges due to their dynamic, conformationally flexible nature, which lacks stable binding pockets typically required for small-molecule . Unlike structured proteins, IDPs adopt transient ensembles of structures, complicating the identification of high-affinity ligands and increasing the risk of off-target effects from promiscuous interactions. This "druggability" issue has historically rendered many IDPs undruggable, as traditional structure-based approaches fail to account for their ensemble-driven functionality. To overcome these hurdles, therapeutic strategies focus on modulating IDP dynamics through stabilizers and degraders. Stabilizers, such as small molecules that induce folding or lock IDPs into non-pathogenic conformations, have shown promise; for instance, compounds like (EGCG) bind to hydrophobic regions of α-synuclein, reducing its conformational fluidity and inhibiting aggregation. Degraders, including proteolysis-targeting chimeras (PROTACs), recruit E3 ligases to ubiquitinate and degrade IDPs via the ubiquitin-proteasome system, bypassing the need for stable binding pockets; recent applications target disordered proteins like and other neurodegeneration-linked IDPs, demonstrating selective degradation in cellular models. These approaches leverage the plasticity of IDPs, turning their disorder into a therapeutic . Recent advances have expanded these strategies, particularly in designing binders and exploiting . In 2025, diffusion-based generative models enabled the design of high-affinity protein binders specific to IDP regions in defined conformations, as demonstrated for targets like and , with dissociation constants in the nanomolar range and validation via . Nanoplatforms, such as engineered nanoparticles, inhibit and Aβ aggregation by sequestering monomeric IDPs and redirecting pathological pathways, reducing neurotoxicity in Alzheimer's models. Additionally, targeting biomolecular condensates—liquid-like assemblies driven by IDP —offers novel interventions in neurodegeneration; condensate-modifying drugs (c-mods) like dissolve aberrant stress granules containing or α-synuclein, restoring cellular . These innovations address gaps in traditional by accommodating IDP ensemble behaviors. Illustrative examples highlight clinical translation. For α-synuclein, implicated in Parkinson's disease, modulators like the monoclonal antibody prasinezumab has advanced to Phase III trials (as of 2025), binding aggregated forms to reduce seeding and progression, with Phase II results showing potential slowed motor decline. As of November 2025, the Phase III PARAISO trial is underway evaluating prasinezumab in early-stage Parkinson's. Similarly, venetoclax, a BH3-mimetic small molecule, targets BCL-2's interaction with intrinsically disordered BH3-only proteins, inhibiting anti-apoptotic signaling in cancers like chronic lymphocytic leukemia; approved by the FDA, it exemplifies how drugs can disrupt IDP-mediated protein-protein interactions, achieving response rates over 70% in relapsed patients.

References

  1. [1]
    Intrinsically Disordered Proteins: An Overview - PubMed Central - NIH
    Nov 14, 2022 · Most acceptably, “intrinsic protein disorder” defines the biologically active proteins or protein segments that exist as ensembles of unfolded, ...
  2. [2]
  3. [3]
    Large-scale Analysis of Thermo-stable, Mammalian Proteins ...
    Based on theoretical translations of whole genome sequences, approximately 30-40% of all eukaryotic proteins are predicted to be either entirely disordered or ...
  4. [4]
    Structure of tumor suppressor p53 and its intrinsically disordered N ...
    As such, with 37% of its structure intrinsically disordered, p53 is typical of the structural content of the human proteome. More than 30% of eukaryotic genomes ...
  5. [5]
    Global Structure of the Intrinsically Disordered Protein Tau Emerges ...
    Mar 1, 2022 · The paradigmatic disordered protein tau plays an important role in neuronal function and neurodegenerative diseases.Introduction · Results and Discussion · Conclusions · Supporting Information
  6. [6]
    Classification of Intrinsically Disordered Regions and Proteins
    Apr 29, 2014 · IDRs and IDPs are prevalent in eukaryotic genomes. For instance, 44% of human protein-coding genes contain disordered segments of >30 amino ...
  7. [7]
    Intrinsically disordered proteins as crucial constituents of cellular ...
    Jan 2, 2015 · For example, IDPs/IDPRs are significantly depleted in a number of order-promoting residues, such as Ile, Leu, Val, Trp, Tyr, Phe, Cys, and Asn, ...
  8. [8]
    Relating sequence encoded information to form and function of ...
    Amino acid compositions of IDPs are characterized by distinct biases [5]. They are deficient in canonical hydrophobic residues and enriched in polar and charged ...
  9. [9]
    Predicting intrinsic disorder in proteins: an overview | Cell Research
    Jul 14, 2009 · A plot of mean net charge versus mean hydropathy (the CH-plot) separates structured and disordered proteins into distinct regions and thereby ...<|control11|><|separator|>
  10. [10]
    Subclassifying disordered proteins by the CH-CDF plot method
    Here we discuss one approach, the (Charge-Hydropathy) versus (Cumulative Distribution Function) plot or CH-CDF plot, which is based the combined use of the CH ...
  11. [11]
    Improving protein order-disorder classification using charge ...
    Dec 16, 2014 · Since entire protein sequences are applied to the original C-H plot by Uversky et al, for consistency, the benchmarking of IDP-Hydropathy scale ...
  12. [12]
    Conformational Entropy of Intrinsically Disordered Proteins ... - Nature
    Jul 3, 2015 · Higher conformational entropy of the intrinsically disordered proteins is also reflected in the less number of two-body contacts, which is ...
  13. [13]
    Distribution of Pico- and Nanosecond Motions in Disordered ... - NIH
    In particular, nuclear spin relaxation can be used to probe a diversity of motions from fast (picoseconds to nanoseconds) reorientation to slow (microseconds to ...
  14. [14]
    Hydrodynamic Radii of Intrinsically Disordered Proteins
    Later work of Zimm included the effect of excluded volume, (67) which resulted in the scaling Rh ∼ Nγ with γ = 0.588. ... radius of gyration Rg and asphericity A.Missing: R_g ~ | Show results with:R_g ~
  15. [15]
    Intrinsically disordered regions are poised to act as sensors of ...
    This sensitivity means IDRs are well suited to act as intracellular sensors, whereby sequence-encoded conformational biases are altered by varying salt, pH, ...
  16. [16]
    Crowder-directed interactions and conformational dynamics in ...
    Dec 21, 2022 · The IDP revealed crowder-specific systematic extension and compaction with the level of macromolecular crowding. Last, a robust extension- ...
  17. [17]
    Intrinsically disordered proteins studied by NMR spectroscopy
    This effect is responsible for narrower NMR lines in IDPs with respect to folded proteins, which partly alleviates the problem of severe cross-peak overlap.
  18. [18]
    Disordered–Ordered Protein Binary Classification by Circular ... - NIH
    Disordered proteins exhibit characteristic CD spectra with an intensive minimum in the vicinity of 200 nm and a low amplitude around 222 nm (Adler et al., 1973; ...
  19. [19]
    A practical guide to small angle X-ray scattering (SAXS) of flexible ...
    Sep 14, 2015 · Small-angle X-ray scattering (SAXS) is a biophysical method to study the overall shape and structural transitions of biological macromolecules in solution.<|control11|><|separator|>
  20. [20]
    Paradoxes and wonders of intrinsic disorder: Prevalence of ... - NIH
    The presence of this gap, where almost all eukaryotes have 32% or more disordered residues, whereas the large majority of the prokaryotic species have 27% or ...Missing: across organisms statistics
  21. [21]
    [PDF] intrinsically disordered proteins: analysis, prediction, simulation, and ...
    ... 45% of human proteins contain long regions that have IDP features, and that, overall, about 45% of the amino acids in humans are located in regions with IDP ...
  22. [22]
    A decade and a half of protein intrinsic disorder: Biology still waits ...
    Uversky VN. Natively unfolded proteins: a point where biology waits for ... The protein trinity—linking function and disorder. Nat Biotechnol. 2001;19 ...
  23. [23]
    improving function annotation of intrinsically disordered proteins
    Oct 30, 2023 · DisProt (URL: https://disprot.org) is the gold standard database for intrinsically disordered proteins and regions, providing valuable information about their ...Abstract · Introduction · Progress and new features · ConclusionsMissing: inception | Show results with:inception
  24. [24]
    IDEAL: Disordered Proteins and Annotations
    IDEAL provides a collection of knowledge of experimentally verified intrinsically disordered proteins (IDPs) and intrinsically disordered regions (IDRs),
  25. [25]
    Abundance of intrinsic disorder in the DNA‐ and RNA‐binding ...
    Apr 1, 2016 · ... Disordered nucleiome: Abundance of intrinsic disorder in the DNA- and RNA-binding proteins in 1121 species from Eukaryota, Bacteria and Archaea.
  26. [26]
    Can proteins be intrinsically disordered inside a membrane? - PMC
    In this hypothesis piece, I argue that intrinsic disorder may exist in membrane embedded proteins, but that it will take a different form due to the different ...Missing: underrepresented | Show results with:underrepresented
  27. [27]
    Evolution of intrinsic disorder in eukaryotic proteins - PMC
    Jun 8, 2017 · Here, we review the evolution of intrinsically disordered regions in eukaryotic proteins. We discuss the interplay between secondary structure and disorder on ...<|control11|><|separator|>
  28. [28]
    Evolutionary analyses of intrinsically disordered regions reveal ...
    We identified signals of widespread constraint, indicating conservation of distributed features is mechanism of IDR evolution common to multiple biological ...
  29. [29]
    Evolution and Disorder - PMC - NIH
    Intrinsically disordered proteins (IDPs) perform essential functions in organisms from viruses to vertebrates, and their improper functioning is responsible for ...
  30. [30]
  31. [31]
  32. [32]
  33. [33]
  34. [34]
  35. [35]
    Functional regulation of an intrinsically disordered protein via a ...
    Jun 28, 2023 · Hence, we next probed the slow dynamics of CytRN using CEST (36) NMR. ... IDPs can access excited conformations with millisecond lifetimes via ...
  36. [36]
    Sequence-Dependent Backbone Dynamics of Intrinsically ...
    Identification of Dynamic Modes in an Intrinsically Disordered Protein Using Temperature-Dependent NMR Relaxation. ... An Arrhenius-type relationship is ...
  37. [37]
    Identification of Dynamic Modes in an Intrinsically Disordered ...
    May 18, 2016 · The dynamic modes and time scales sampled by intrinsically disordered proteins (IDPs) define their function.
  38. [38]
    Chain dynamics of unfolded and intrinsically disordered proteins ...
    Since the reconfiguration of unfolded proteins in the absence of persistent interactions occurs on the sub-microsecond time scale, MD simulations can now access ...
  39. [39]
    Folding Free Energy Landscape of Ordered and Intrinsically ...
    Oct 17, 2019 · We demonstrate that HP-35 and WW domain indeed exhibit the steep folding funnel: the landscape slope for these proteins is ca. -50 kcal/mol.
  40. [40]
    Structural and thermodynamic analyses of the β-to-α transformation ...
    Oct 18, 2022 · IDPs, in contrast, exhibit a rather flat energy landscape and no specific conformation is favored, that is, significantly populated. Fold- ...<|control11|><|separator|>
  41. [41]
    Differential scanning microcalorimetry of intrinsically disordered ...
    The thermal dependence of protein heat capacity extracted from DSC data is a valuable source of information on intrinsic disorder level of a protein.
  42. [42]
    Intrinsic disorder as a mechanism to optimize allosteric coupling in ...
    May 15, 2007 · Here, we present such a model and show that site-to-site allosteric coupling is maximized when intrinsic disorder is present in the domains or ...
  43. [43]
    Intrinsically Disordered Proteins and Allosteric Regulation - PMC
    Allostery within or mediated by intrinsically disordered proteins ensures robust and efficient signal integration through mechanisms that would be extremely ...
  44. [44]
  45. [45]
    Linking Folding and Binding - PMC - NIH
    Linking folding and binding refers to the coupling of folding of intrinsically disordered proteins with binding to a target molecule, often involving short ...
  46. [46]
    Structure of the p53 transactivation domain in complex with the ...
    NMR studies revealed that the free p53 TAD has a weak propensity to form transient helical structure between residues 18 and 26 and two turns (residues 40–44 ...
  47. [47]
    Consequences of Fuzziness in the NF-κB/IκBκ Interaction - PMC
    This chapter provides a short review of various biophysical experiments that have been applied to the inhibitor of kappa B, IκBα, and its binding partner.Missing: IDP | Show results with:IDP
  48. [48]
    Accurate model of liquid–liquid phase behavior of intrinsically ...
    Many intrinsically disordered proteins (IDPs) may undergo liquid–liquid phase separation (LLPS) and participate in the formation of membraneless organelles ...
  49. [49]
    FUS and TDP-43 Phases in Health and Disease - PMC
    The distinct prion-like domains (PrLDs) of FUS and TDP-43, modulate phase transitions that result in condensates with a range of material states.
  50. [50]
    Liquid-Liquid Phase Separation of TDP-43 and FUS in Physiology ...
    In this article, we review the factors that mediate and regulate phase separation of TDP-43 and FUS. We also review evidences that connect the phase separation ...Abstract · Introduction · Factors That Mediate Liquid... · Functional Role of Phase...Missing: IDP | Show results with:IDP
  51. [51]
    Liquid phase condensation in cell physiology and disease - Science
    Sep 22, 2017 · Liquid phase condensation increasingly appears to be a fundamental mechanism for organizing intracellular space.Phase Separation And... · Liquid Droplets In The Cell · Liquid Metastability And...<|control11|><|separator|>
  52. [52]
    Phase Separation of Intrinsically Disordered Nucleolar Proteins ...
    Dec 3, 2021 · In this review, we discuss the nucleolus as an example of the establishment, dynamics, and maintenance of a membraneless organelle with a high level of ...
  53. [53]
    Recent trends in studies of biomolecular phase separation.
    Recently, phase separation of TDP-43 has been investigated in vitro, using CTD sequence variants. The mutants showed weaker binding affinity to another binding ...
  54. [54]
    Cross-Talk of Cation−π Interactions with Electrostatic and Aromatic ...
    Sep 18, 2023 · We found greater stabilization of up to 80% via cation−π interactions in condensates formed from peptides with higher aromatic residue content or less charge ...Supporting Information · Author Information · References
  55. [55]
    Sequence-based prediction of intermolecular interactions driven by ...
    May 22, 2025 · This method provides a rapid way to characterize IDRs directly from sequence and identify and prioritize potential cellular interactions for experimental ...
  56. [56]
    Experimental methods to study the structure and dynamics of ...
    Its sensitivity to changes in the protein environment such as the pH, temperature, and ligand binding allows the study of effects of various conditions on IDPs.
  57. [57]
    NMR Provides Unique Insight into the Functional Dynamics and ...
    Apr 21, 2022 · We have validated the results against NMR spectroscopy and SAXS measurements, obtaining reasonable agreement. The two helixes which most ...
  58. [58]
    Structural Proteomics Methods to Interrogate the Conformations and ...
    Mar 10, 2021 · NMR is the most often used method to study IDPs as it can provide a signature for intrinsic disorder (low 1H and 15N amide chemical shift ...
  59. [59]
    Single Molecule FRET: A Powerful Tool to Study Intrinsically ... - MDPI
    Nov 8, 2018 · Single molecule and ensemble FRET measurements combined with other methods including NMR and EPR techniques will provide valuable benchmarks ...Single Molecule Fret: A... · 1. Introduction · 4.2. Phosphorylation...<|separator|>
  60. [60]
    (PDF) How Random are Intrinsically Disordered Proteins? A Small ...
    Aug 6, 2025 · Small-angle X-ray and neutron scattering (SAXS and SANS) are fundamental tools used to study the global shapes of proteins, nucleic acids ...
  61. [61]
    Sedimentation velocity analytical ultracentrifugation for intrinsically ...
    This chapter is a step-by-step protocol for setting up, executing and analyzing SV experiments in the context of the characterization of IDPs, based on a real ...
  62. [62]
    Structural biases in disordered proteins are prevalent in the cell - PMC
    Here we show that structural biases in IDP ensembles found in vitro are recapitulated inside human-derived cells.
  63. [63]
    High-Throughput Screening Methodology to Identify Alpha ... - MDPI
    The lack of reproducibility between aggregation curves is a strong limitation to identify bona fide aggregation inhibitors, since their potency becomes hidden ...
  64. [64]
    Recent Force Field Strategies for Intrinsically Disordered Proteins
    This review covers recent force field strategies proposed for more accurate molecular dynamics simulations of IDPs. The strategies include adjusting dihedral ...
  65. [65]
    Atomistic molecular dynamics simulations of intrinsically disordered ...
    All-atom MD simulations are now explaining the sequence-dependent dynamics of IDPs; elucidating the mechanisms of their binding to other proteins, nucleic acids ...
  66. [66]
    ff14IDPs Force Field Improving the Conformation Sampling of ... - NIH
    In order to improve the accuracy of conformational sampling for structural proteins, the Amber ff14SB force field was recently developed to refine the protein ...
  67. [67]
    AWSEM-IDP: A Coarse-Grained Force Field for Intrinsically ... - NIH
    In this work, we introduce AWSEM-IDP, a new AWSEM branch for simulating intrinsically disordered proteins (IDPs), where the weights of the potentials ...
  68. [68]
    Full structural ensembles of intrinsically disordered proteins from ...
    Feb 23, 2021 · The IDPs we studied mostly showed a high propensity for coils that lack secondary structure, consistent with the lack of long-range contacts ...
  69. [69]
    Generation of the configurational ensemble of an intrinsically ...
    Sep 23, 2019 · We report that enhancing the sampling of the simulations can generate an ensemble of IDP structures in quantitative agreement with scattering and NMR.Missing: seminal | Show results with:seminal
  70. [70]
    Molecular Dynamics Simulations Combined with Nuclear Magnetic ...
    Mar 6, 2019 · We survey some recent studies combining MD simulations with NMR and/or SAXS measurements to investigate the relationship between IDP conformational ensemble ...
  71. [71]
    Molecular Simulations Integrated with Experiments for Probing the ...
    Dec 19, 2023 · Advances in tailoring molecular simulations for IDPs combined with experimental cross-validation offer an atomistic view of the mechanisms that control IDP ...
  72. [72]
    Towards Accurate Simulation of Coupling between Protein ...
    Jan 10, 2025 · Intrinsically disordered proteins (IDPs) frequently mediate phase separation that underlies the formation of biomolecular condensate.
  73. [73]
    Backbone interactions and secondary structures in phase ...
    Feb 13, 2024 · We further focus on recent advances in experimental techniques and molecular dynamics simulation methods for probing and exploring the roles of ...<|separator|>
  74. [74]
  75. [75]
  76. [76]
    AlphaFold and Implications for Intrinsically Disordered Proteins
    Intrinsically disordered regions often evolve more rapidly than ordered regions and thus alignments of these regions are generally poorer and involve large gaps ...
  77. [77]
    The power and pitfalls of AlphaFold2 for structure prediction beyond ...
    IDPs represents a challenge for AF2 since it is difficult to identify evolutionary constraints from MSAs of IDPs and IDRs due to sequence hypervariability.
  78. [78]
    Before and after AlphaFold2: An overview of protein structure ...
    Feb 27, 2023 · ... protein structures, AlphaFold2 models have important limitations. First, AlphaFold2 has difficulty to predict intrinsically disordered proteins ...
  79. [79]
    AlphaFold prediction of structural ensembles of disordered proteins
    Feb 14, 2025 · PED in 2021: a major update of the protein ensemble database for intrinsically disordered proteins. Nucleic Acids Res. 49, D404–D411 (2021) ...
  80. [80]
    Prediction of Structural Ensembles of Disordered Proteins - bioRxiv
    Nov 10, 2024 · We introduce the AlphaFold-Metainference method to use AlphaFold-derived distances as structural restraints in molecular dynamics simulations.
  81. [81]
    IDP-LM: Prediction of protein intrinsic disorder ... - Research journals
    Nov 22, 2023 · In this study, we proposed a computational predictor called IDP-LM for predicting intrinsic disorder and disorder functions by leveraging the pre-trained ...
  82. [82]
    Deep Learning for Hyperpolarized NMR of Intrinsically Disordered ...
    Aug 8, 2025 · This approach is based on a first-principles understanding of dDNP line shapes and delivers a scalable solution to spectral distortion. Applied ...1 Introduction · 2.1 Data Treatment... · 2.2 Osteopontin
  83. [83]
    Diffusing protein binders to intrinsically disordered proteins - Nature
    Jul 30, 2025 · Proteins that bind to intrinsically disordered proteins (IDPs) and intrinsically disordered regions (IDRs) with high affinity and ...
  84. [84]
    ELM—the Eukaryotic Linear Motif resource—2024 update
    These methods enable the discovery of large numbers of candidate SLiMs in the intrinsically disordered proteomes of hosts and pathogens that can be followed up ...
  85. [85]
    Release notes - DisProt
    more than 600 entries (19%) have MIADE information. Version 9.5 − December 2023. Annotation release 2023_12 (narrative blogpost ). New dataset: condensates ...Missing: 9 | Show results with:9
  86. [86]
    Prediction of Protein Binding Regions in Disordered Proteins
    We report ANCHOR, a method for the prediction of disordered binding regions. ANCHOR relies on the pairwise energy estimation approach that is the basis of ...
  87. [87]
    MOBIDB in 2025: integrating ensemble properties and function ...
    Oct 29, 2024 · The number of processed proteins from PDB data is 65 290 corresponding to 216 599 PDB entries and 731 924 different chains, representing an ...
  88. [88]
    The Sherpa hypothesis: Phenotype-Preserving Disordered Proteins ...
    Jul 11, 2023 · IDPs are frequently implicated in various maladies, such as cancer and neurodegenerative diseases. For example, mutations in the disordered ...Missing: review | Show results with:review
  89. [89]
    p53 Proteoforms and Intrinsic Disorder: An Illustration of the Protein ...
    This protein has numerous biological functions, possesses intrinsically disordered regions crucial for its functionality, can form both homo-tetramers and ...
  90. [90]
    Intrinsically Disordered Proteins and Human Diseases
    Shown are the percentages of predicted order transitions for different classes of mutation in intrinsically disordered regions (IDPRs, left) and ordered regions ...
  91. [91]
  92. [92]
    Proteins with Intrinsically Disordered Domains Are Preferentially ...
    Aug 28, 2015 · Our study indicates that neurodegenerative disease-associated proteins are particularly vulnerable to recruitment into polyQ aggregates via their ID domains.
  93. [93]
    Membraneless organelles in health and disease - Nature
    Nov 18, 2024 · From neurodegenerative disorders, cardiovascular diseases, aging, and metabolism to cancer, the influence of MLOs on human health and disease ...
  94. [94]
    How to drug a cloud? Targeting intrinsically disordered proteins
    Nov 21, 2024 · This article describes the major structural features of IDPs/IDRs and the peculiarities of the disorder-based functionality. It also discusses ...
  95. [95]
    Fuzzy Drug Targets: Disordered Proteins in the Drug-Discovery Realm
    Mar 7, 2023 · A review on proteins ... Intrinsically disordered proteins are attractive therapeutic targets owing to their prevalence in several diseases.Introduction · Experimental Techniques in... · IDPs Involved in Disease...<|separator|>
  96. [96]
    Structure-based Inhibitor Design for the Intrinsically Disordered ...
    Mar 2, 2016 · Here, we describe a novel computational approach to virtually screen for compounds that can simultaneously bind to different IDP conformations.<|separator|>
  97. [97]
    Current perspectives in drug targeting intrinsically disordered ...
    May 6, 2025 · This review explores current strategies to target IDPs and biomolecular condensates, potential improvements, and the challenges and opportunities in this ...
  98. [98]
    Exploring Degradation of Intrinsically Disordered Protein YAP ...
    Sep 22, 2023 · A novel approach utilizing Proteolysis-Targeting Chimera (PROTAC) technology was employed to design and synthesize a series of YAP degraders.
  99. [99]
    Degradation of intrinsically disordered proteins using proteolysis ...
    Nov 28, 2024 · For instance, a PROTAC typically carries a small molecule ligand to bind with E3 ligase and another ligand to recruit a target protein.
  100. [100]
    Nanoplatforms Targeting Intrinsically Disordered Protein ...
    May 8, 2025 · In this review, we explore diverse multifunctional nanoplatform strategies targeting and sequestering pathological IDPs, including tau, Aβ, and ...
  101. [101]
    The roles of intrinsically disordered proteins in neurodegeneration
    Proteins like Tau, α-synuclein, FUS, and TDP-43 undergo phase separation, which can lead to pathological aggregation and the formation of insoluble deposits ...
  102. [102]
    An update on immune-based alpha-synuclein trials in Parkinson's ...
    Dec 12, 2024 · This review provides a comprehensive analysis of recent clinical trials involving vaccines and monoclonal antibodies targeting α-syn.Missing: IDPs | Show results with:IDPs
  103. [103]
    Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic ...
    Dec 6, 2015 · Selective targeting of BCL2 with venetoclax had a manageable safety profile and induced substantial responses in patients with relapsed CLL or SLL.Missing: binding disordered IDP