fbpx
Wikipedia

Intrinsically disordered proteins

In molecular biology, an intrinsically disordered protein (IDP) is a protein that lacks a fixed or ordered three-dimensional structure,[2][3][4] typically in the absence of its macromolecular interaction partners, such as other proteins or RNA. IDPs range from fully unstructured to partially structured and include random coil, molten globule-like aggregates, or flexible linkers in large multi-domain proteins. They are sometimes considered as a separate class of proteins along with globular, fibrous and membrane proteins.[5]

Conformational flexibility in SUMO-1 protein (PDB:1a5r). The central part shows relatively ordered structure. Conversely, the N- and C-terminal regions (left and right, respectively) show ‘intrinsic disorder’, although a short helical region persists in the N-terminal tail. Ten alternative NMR models were morphed. Secondary structure elements: α-helices (red), β-strands (blue arrows). [1]

IDPs are a very large and functionally important class of proteins and their discovery has disproved the idea that three-dimensional structures of proteins must be fixed to accomplish their biological functions. For example, IDPs have been identified to participate in weak multivalent interactions that are highly cooperative and dynamic, lending them importance in DNA regulation and in cell signaling.[6][7] Many IDPs can also adopt a fixed three-dimensional structure after binding to other macromolecules. Overall, IDPs are different from structured proteins in many ways and tend to have distinctive function, structure, sequence, interactions, evolution and regulation.[8]

History edit

 
An ensemble of NMR structures of the Thylakoid soluble phosphoprotein TSP9, which shows a largely flexible protein chain.[9]

In the 1930s-1950s, the first protein structures were solved by protein crystallography. These early structures suggested that a fixed three-dimensional structure might be generally required to mediate biological functions of proteins. These publications solidified the central dogma of molecular biology in that the amino acid sequence of a protein determines its structure which, in turn, determines its function. In 1950, Karush wrote about 'Configurational Adaptability' contradicting this assumption. He was convinced that proteins have more than one configuration at the same energy level and can choose one when binding to other substrates. In the 1960s, Levinthal's paradox suggested that the systematic conformational search of a long polypeptide is unlikely to yield a single folded protein structure on biologically relevant timescales (i.e. microseconds to minutes). Curiously, for many (small) proteins or protein domains, relatively rapid and efficient refolding can be observed in vitro. As stated in Anfinsen's Dogma from 1973, the fixed 3D structure of these proteins is uniquely encoded in its primary structure (the amino acid sequence), is kinetically accessible and stable under a range of (near) physiological conditions, and can therefore be considered as the native state of such "ordered" proteins.[10]

During the subsequent decades, however, many large protein regions could not be assigned in x-ray datasets, indicating that they occupy multiple positions, which average out in electron density maps. The lack of fixed, unique positions relative to the crystal lattice suggested that these regions were "disordered". Nuclear magnetic resonance spectroscopy of proteins also demonstrated the presence of large flexible linkers and termini in many solved structural ensembles.

In 2001, Dunker questioned whether the newly found information was ignored for 50 years[11] with more quantitative analyses becoming available in the 2000s.[12] In the 2010s it became clear that IDPs are common among disease-related proteins, such as alpha-synuclein and tau.[13]

Abundance edit

It is now generally accepted that proteins exist as an ensemble of similar structures with some regions more constrained than others. IDPs occupy the extreme end of this spectrum of flexibility and include proteins of considerable local structure tendency or flexible multidomain assemblies.[14][15]

Intrinsic disorder is particularly elevated among proteins that regulate chromatin and transcription,[16] and bioinformatic predictions indicate that is more common in genomes and proteomes than in known structures in the protein database. Based on DISOPRED2 prediction, long (>30 residue) disordered segments occur in 2.0% of archaean, 4.2% of eubacterial and 33.0% of eukaryotic proteins,[12] including certain disease-related proteins.[13]

Biological roles edit

Highly dynamic disordered regions of proteins have been linked to functionally important phenomena such as allosteric regulation and enzyme catalysis.[14][15] Many disordered proteins have the binding affinity with their receptors regulated by post-translational modification, thus it has been proposed that the flexibility of disordered proteins facilitates the different conformational requirements for binding the modifying enzymes as well as their receptors.[17] Intrinsic disorder is particularly enriched in proteins implicated in cell signaling and transcription,[16] as well as chromatin remodeling functions.[18][19] Genes that have recently been born de novo tend to have higher disorder.[20][21]

Flexible linkers edit

Disordered regions are often found as flexible linkers or loops connecting domains. Linker sequences vary greatly in length but are typically rich in polar uncharged amino acids. Flexible linkers allow the connecting domains to freely twist and rotate to recruit their binding partners via protein domain dynamics. They also allow their binding partners to induce larger scale conformational changes by long-range allostery.[14][2] The flexible linker of FBP25 which connects two domains of FKBP25 is important for the binding of FKBP25 with DNA.[22]

Linear motifs edit

Linear motifs are short disordered segments of proteins that mediate functional interactions with other proteins or other biomolecules (RNA, DNA, sugars etc.).[16] Many roles of linear motifs are associated with cell regulation, for instance in control of cell shape, subcellular localisation of individual proteins and regulated protein turnover. Often, post-translational modifications such as phosphorylation tune the affinity (not rarely by several orders of magnitude) of individual linear motifs for specific interactions. Relatively rapid evolution and a relatively small number of structural restraints for establishing novel (low-affinity) interfaces make it particularly challenging to detect linear motifs but their widespread biological roles and the fact that many viruses mimick/hijack linear motifs to efficiently recode infected cells underlines the timely urgency of research on this very challenging and exciting topic.

Pre-structured motifs edit

Unlike globular proteins, IDPs do not have spatially-disposed active pockets. Fascinatingly, 80% of target-unbound IDPs (~4 dozens) subjected to detailed structural characterization by NMR possess linear motifs termed PresMos (pre-structured motifs) [23] that are transient secondary structural elements primed for target recognition. In several cases it has been demonstrated that these transient structures become full and stable secondary structures, e.g., helices, upon target binding. Hence, PresMos are the putative active sites in IDPs.

Coupled folding and binding edit

Many unstructured proteins undergo transitions to more ordered states upon binding to their targets (e.g. Molecular Recognition Features (MoRFs)[24]). The coupled folding and binding may be local, involving only a few interacting residues, or it might involve an entire protein domain. It was recently shown that the coupled folding and binding allows the burial of a large surface area that would be possible only for fully structured proteins if they were much larger.[25] Moreover, certain disordered regions might serve as "molecular switches" in regulating certain biological function by switching to ordered conformation upon molecular recognition like small molecule-binding, DNA/RNA binding, ion interactions etc.[26]

The ability of disordered proteins to bind, and thus to exert a function, shows that stability is not a required condition. Many short functional sites, for example Short Linear Motifs are over-represented in disordered proteins. Disordered proteins and short linear motifs are particularly abundant in many RNA viruses such as Hendra virus, HCV, HIV-1 and human papillomaviruses. This enables such viruses to overcome their informationally limited genomes by facilitating binding, and manipulation of, a large number of host cell proteins.[27][28]

Disorder in the bound state (fuzzy complexes) edit

Intrinsically disordered proteins can retain their conformational freedom even when they bind specifically to other proteins. The structural disorder in bound state can be static or dynamic. In fuzzy complexes structural multiplicity is required for function and the manipulation of the bound disordered region changes activity. The conformational ensemble of the complex is modulated via post-translational modifications or protein interactions.[29] Specificity of DNA binding proteins often depends on the length of fuzzy regions, which is varied by alternative splicing.[30] Some fuzzy complexes may exhibit high binding affinity,[31] although other studies showed different affinity values for the same system in a different concentration regime.[32]

Structural aspects edit

Intrinsically disordered proteins adapt many different structures in vivo according to the cell's conditions, creating a structural or conformational ensemble.[33][34]

Therefore, their structures are strongly function-related. However, only few proteins are fully disordered in their native state. Disorder is mostly found in intrinsically disordered regions (IDRs) within an otherwise well-structured protein. The term intrinsically disordered protein (IDP) therefore includes proteins that contain IDRs as well as fully disordered proteins.

The existence and kind of protein disorder is encoded in its amino acid sequence.[2] In general, IDPs are characterized by a low content of bulky hydrophobic amino acids and a high proportion of polar and charged amino acids, usually referred to as low hydrophobicity.[33] This property leads to good interactions with water. Furthermore, high net charges promote disorder because of electrostatic repulsion resulting from equally charged residues.[34] Thus disordered sequences cannot sufficiently bury a hydrophobic core to fold into stable globular proteins. In some cases, hydrophobic clusters in disordered sequences provide the clues for identifying the regions that undergo coupled folding and binding (refer to biological roles). Many disordered proteins reveal regions without any regular secondary structure. These regions can be termed as flexible, compared to structured loops. While the latter are rigid and contain only one set of Ramachandran angles, IDPs involve multiple sets of angles.[34] The term flexibility is also used for well-structured proteins, but describes a different phenomenon in the context of disordered proteins. Flexibility in structured proteins is bound to an equilibrium state, while it is not so in IDPs.[34] Many disordered proteins also reveal low complexity sequences, i.e. sequences with over-representation of a few residues. While low complexity sequences are a strong indication of disorder, the reverse is not necessarily true, that is, not all disordered proteins have low complexity sequences. Disordered proteins have a low content of predicted secondary structure.

Due to the disordered nature of these proteins, topological approaches have been developed to search for conformational patterns in their dynamics. For instance, circuit topology has been applied to track the dynamics of disordered protein domains.[35] By employing a topological approach, one can categorize motifs according to their topological buildup and the timescale of their formation.

Experimental validation edit

IDPs can be validated in several contexts. Most approaches for experimental validation of IDPs are restricted to extracted or purified proteins while some new experimental strategies aim to explore in vivo conformations and structural variations of IDPs inside intact living cells and systematic comparisons between their dynamics in vivo and in vitro.

In vivo approaches edit

The first direct evidence for in vivo persistence of intrinsic disorder has been achieved by in-cell NMR upon electroporation of a purified IDP and recovery of cells to an intact state.[36]

Larger-scale in vivo validation of IDR predictions is now possible using biotin 'painting'.[37][38]

In vitro approaches edit

Intrinsically unfolded proteins, once purified, can be identified by various experimental methods. The primary method to obtain information on disordered regions of a protein is NMR spectroscopy. The lack of electron density in X-ray crystallographic studies may also be a sign of disorder.

Folded proteins have a high density (partial specific volume of 0.72-0.74 mL/g) and commensurately small radius of gyration. Hence, unfolded proteins can be detected by methods that are sensitive to molecular size, density or hydrodynamic drag, such as size exclusion chromatography, analytical ultracentrifugation, small angle X-ray scattering (SAXS), and measurements of the diffusion constant. Unfolded proteins are also characterized by their lack of secondary structure, as assessed by far-UV (170-250 nm) circular dichroism (esp. a pronounced minimum at ~200 nm) or infrared spectroscopy. Unfolded proteins also have exposed backbone peptide groups exposed to solvent, so that they are readily cleaved by proteases, undergo rapid hydrogen-deuterium exchange and exhibit a small dispersion (<1 ppm) in their 1H amide chemical shifts as measured by NMR. (Folded proteins typically show dispersions as large as 5 ppm for the amide protons.) Recently, new methods including Fast parallel proteolysis (FASTpp) have been introduced, which allow to determine the fraction folded/disordered without the need for purification.[39][40] Even subtle differences in the stability of missense mutations, protein partner binding and (self)polymerisation-induced folding of (e.g.) coiled-coils can be detected using FASTpp as recently demonstrated using the tropomyosin-troponin protein interaction.[41] Fully unstructured protein regions can be experimentally validated by their hypersusceptibility to proteolysis using short digestion times and low protease concentrations.[42]

Bulk methods to study IDP structure and dynamics include SAXS for ensemble shape information, NMR for atomistic ensemble refinement, Fluorescence for visualising molecular interactions and conformational transitions, x-ray crystallography to highlight more mobile regions in otherwise rigid protein crystals, cryo-EM to reveal less fixed parts of proteins, light scattering to monitor size distributions of IDPs or their aggregation kinetics, NMR chemical shift and Circular Dichroism to monitor secondary structure of IDPs.

Single-molecule methods to study IDPs include spFRET[43] to study conformational flexibility of IDPs and the kinetics of structural transitions, optical tweezers[44] for high-resolution insights into the ensembles of IDPs and their oligomers or aggregates, nanopores[45] to reveal global shape distributions of IDPs, magnetic tweezers[46] to study structural transitions for long times at low forces, high-speed AFM[47] to visualise the spatio-temporal flexibility of IDPs directly.

Disorder annotation edit

 
REMARK465 - missing electron densities in X-ray structure representing protein disorder (PDB: 1a22​, human growth hormone bound to receptor). Compilation of screenshots from PDB database and molecule representation via VMD. Blue and red arrows point to missing residues on receptor and growth hormone, respectively.

Intrinsic disorder can be either annotated from experimental information or predicted with specialized software. Disorder prediction algorithms can predict Intrinsic Disorder (ID) propensity with high accuracy (approaching around 80%) based on primary sequence composition, similarity to unassigned segments in protein x-ray datasets, flexible regions in NMR studies and physico-chemical properties of amino acids.

Disorder databases edit

Databases have been established to annotate protein sequences with intrinsic disorder information. The DisProt database contains a collection of manually curated protein segments which have been experimentally determined to be disordered. MobiDB is a database combining experimentally curated disorder annotations (e.g. from DisProt) with data derived from missing residues in X-ray crystallographic structures and flexible regions in NMR structures.

Predicting IDPs by sequence edit

Separating disordered from ordered proteins is essential for disorder prediction. One of the first steps to find a factor that distinguishes IDPs from non-IDPs is to specify biases within the amino acid composition. The following hydrophilic, charged amino acids A, R, G, Q, S, P, E and K have been characterized as disorder-promoting amino acids, while order-promoting amino acids W, C, F, I, Y, V, L, and N are hydrophobic and uncharged. The remaining amino acids H, M, T and D are ambiguous, found in both ordered and unstructured regions.[2] A more recent analysis ranked amino acids by their propensity to form disordered regions as follows (order promoting to disorder promoting): W, F, Y, I, M, L, V, N, C, T, A, G, R, D, H, Q, K, S, E, P.[48] As it can be seen from the list, small, charged, hydrophilic residues often promote disorder, while large and hydrophobic residues promote order.

This information is the basis of most sequence-based predictors. Regions with little to no secondary structure, also known as NORS (NO Regular Secondary structure) regions,[49] and low-complexity regions can easily be detected. However, not all disordered proteins contain such low complexity sequences.

Prediction methods edit

Determining disordered regions from biochemical methods is very costly and time-consuming. Due to the variable nature of IDPs, only certain aspects of their structure can be detected, so that a full characterization requires a large number of different methods and experiments. This further increases the expense of IDP determination. In order to overcome this obstacle, computer-based methods are created for predicting protein structure and function. It is one of the main goals of bioinformatics to derive knowledge by prediction. Predictors for IDP function are also being developed, but mainly use structural information such as linear motif sites.[4][50] There are different approaches for predicting IDP structure, such as neural networks or matrix calculations, based on different structural and/or biophysical properties.

Many computational methods exploit sequence information to predict whether a protein is disordered.[51] Notable examples of such software include IUPRED and Disopred. Different methods may use different definitions of disorder. Meta-predictors show a new concept, combining different primary predictors to create a more competent and exact predictor.

Due to the different approaches of predicting disordered proteins, estimating their relative accuracy is fairly difficult. For example, neural networks are often trained on different datasets. The disorder prediction category is a part of biannual CASP experiment that is designed to test methods according accuracy in finding regions with missing 3D structure (marked in PDB files as REMARK465, missing electron densities in X-ray structures).

Disorder and disease edit

Intrinsically unstructured proteins have been implicated in a number of diseases.[13] Aggregation of misfolded proteins is the cause of many synucleinopathies and toxicity as those proteins start binding to each other randomly and can lead to cancer or cardiovascular diseases. Thereby, misfolding can happen spontaneously because millions of copies of proteins are made during the lifetime of an organism. The aggregation of the intrinsically unstructured protein α-synuclein is thought to be responsible. The structural flexibility of this protein together with its susceptibility to modification in the cell leads to misfolding and aggregation. Genetics, oxidative and nitrative stress as well as mitochondrial impairment impact the structural flexibility of the unstructured α-synuclein protein and associated disease mechanisms.[52] Many key tumour suppressors have large intrinsically unstructured regions, for example p53 and BRCA1. These regions of the proteins are responsible for mediating many of their interactions. Taking the cell's native defense mechanisms as a model drugs can be developed, trying to block the place of noxious substrates and inhibiting them, and thus counteracting the disease.[53]

Computer simulations edit

 
MD simulation of the Glutaredoxin 1 from Trypanosoma Brucei. The globular thioredoxin fold is depicted in blue, while the disordered N-tail in green. According to the MD results, the disordered tail can be modulating the dynamics of the binding pocket.[54]

Owing to high structural heterogeneity, NMR/SAXS experimental parameters obtained will be an average over a large number of highly diverse and disordered states (an ensemble of disordered states). Hence, to understand the structural implications of these experimental parameters, there is a necessity for accurate representation of these ensembles by computer simulations. All-atom molecular dynamic simulations can be used for this purpose but their use is limited by the accuracy of current force-fields in representing disordered proteins. Nevertheless, some force-fields have been explicitly developed for studying disordered proteins by optimising force-field parameters using available NMR data for disordered proteins. (examples are CHARMM 22*, CHARMM 32,[55] Amber ff03* etc.)

MD simulations restrained by experimental parameters (restrained-MD) have also been used to characterise disordered proteins.[56][57][58] In principle, one can sample the whole conformational space given an MD simulation (with accurate Force-field) is run long enough. Because of very high structural heterogeneity, the time scales that needs to be run for this purpose are very large and are limited by computational power. However, other computational techniques such as accelerated-MD simulations,[59] replica exchange simulations,[60]

[61] metadynamics,[62][63] multicanonical MD simulations,[64] or methods using coarse-grained representation with implicit and explicit solvents[65][66][67] have been used to sample broader conformational space in smaller time scales.

Moreover, various protocols and methods of analyzing IDPs, such as studies based on quantitative analysis of GC content in genes and their respective chromosomal bands, have been used to understand functional IDP segments.[68][69]

See also edit

References edit

  1. ^ Majorek K, Kozlowski L, Jakalski M, Bujnicki JM (December 18, 2008). "First Steps of Protein Structure Prediction" (PDF). In Bujnicki J (ed.). Prediction of Protein Structures, Functions, and Interactions. John Wiley & Sons, Ltd. pp. 39–62. doi:10.1002/9780470741894.ch2. ISBN 9780470517673.
  2. ^ a b c d Dunker AK, Lawson JD, Brown CJ, Williams RM, Romero P, Oh JS, Oldfield CJ, Campen AM, Ratliff CM, Hipps KW, Ausio J, Nissen MS, Reeves R, Kang C, Kissinger CR, Bailey RW, Griswold MD, Chiu W, Garner EC, Obradovic Z (2001). "Intrinsically disordered protein". Journal of Molecular Graphics & Modelling. 19 (1): 26–59. CiteSeerX 10.1.1.113.556. doi:10.1016/s1093-3263(00)00138-8. PMID 11381529.
  3. ^ Dyson HJ, Wright PE (March 2005). "Intrinsically unstructured proteins and their functions". Nature Reviews Molecular Cell Biology. 6 (3): 197–208. doi:10.1038/nrm1589. PMID 15738986. S2CID 18068406.
  4. ^ a b Dunker AK, Silman I, Uversky VN, Sussman JL (December 2008). "Function and structure of inherently disordered proteins". Current Opinion in Structural Biology. 18 (6): 756–64. doi:10.1016/j.sbi.2008.10.002. PMID 18952168.
  5. ^ Andreeva A, Howorth D, Chothia C, Kulesha E, Murzin AG (January 2014). "SCOP2 prototype: a new approach to protein structure mining". Nucleic Acids Research. 42 (Database issue): D310–4. doi:10.1093/nar/gkt1242. PMC 3964979. PMID 24293656.
  6. ^ Mir M, Stadler MR, Ortiz SA, Hannon CE, Harrison MM, Darzacq X, Eisen MB (December 2018). Singer RH, Struhl K, Crocker J (eds.). "Dynamic multifactor hubs interact transiently with sites of active transcription in Drosophila embryos". eLife. 7: e40497. doi:10.7554/eLife.40497. PMC 6307861. PMID 30589412.
  7. ^ Wright PE, Dyson HJ (January 2015). "Intrinsically disordered proteins in cellular signalling and regulation". Nature Reviews. Molecular Cell Biology. 16 (1): 18–29. doi:10.1038/nrm3920. PMC 4405151. PMID 25531225.
  8. ^ van der Lee R, Buljan M, Lang B, Weatheritt RJ, Daughdrill GW, Dunker AK, et al. (July 2014). "Classification of intrinsically disordered regions and proteins". Chemical Reviews. 114 (13): 6589–6631. doi:10.1021/cr400525m. PMC 4095912. PMID 24773235.
  9. ^ Song J, Lee MS, Carlberg I, Vener AV, Markley JL (December 2006). "Micelle-induced folding of spinach thylakoid soluble phosphoprotein of 9 kDa and its functional implications". Biochemistry. 45 (51): 15633–43. doi:10.1021/bi062148m. PMC 2533273. PMID 17176085.
  10. ^ Anfinsen CB (July 1973). "Principles that govern the folding of protein chains". Science. 181 (4096): 223–230. Bibcode:1973Sci...181..223A. doi:10.1126/science.181.4096.223. PMID 4124164.
  11. ^ Dunker AK, Lawson JD, Brown CJ, Williams RM, Romero P, Oh JS, Oldfield CJ, Campen AM, Ratliff CM, Hipps KW, Ausio J, Nissen MS, Reeves R, Kang C, Kissinger CR, Bailey RW, Griswold MD, Chiu W, Garner EC, Obradovic Z (2001-01-01). "Intrinsically disordered protein". Journal of Molecular Graphics & Modelling. 19 (1): 26–59. CiteSeerX 10.1.1.113.556. doi:10.1016/s1093-3263(00)00138-8. PMID 11381529.
  12. ^ a b Ward JJ, Sodhi JS, McGuffin LJ, Buxton BF, Jones DT (March 2004). "Prediction and functional analysis of native disorder in proteins from the three kingdoms of life". Journal of Molecular Biology. 337 (3): 635–45. CiteSeerX 10.1.1.120.5605. doi:10.1016/j.jmb.2004.02.002. PMID 15019783.
  13. ^ a b c Uversky VN, Oldfield CJ, Dunker AK (2008). "Intrinsically disordered proteins in human diseases: introducing the D2 concept". Annual Review of Biophysics. 37: 215–46. doi:10.1146/annurev.biophys.37.032807.125924. PMID 18573080.
  14. ^ a b c Bu Z, Callaway DJ (2011). "Proteins MOVE! Protein dynamics and long-range allostery in cell signaling". Protein Structure and Diseases. Advances in Protein Chemistry and Structural Biology. Vol. 83. pp. 163–221. doi:10.1016/B978-0-12-381262-9.00005-7. ISBN 9780123812629. PMID 21570668.
  15. ^ a b Kamerlin SC, Warshel A (May 2010). "At the dawn of the 21st century: Is dynamics the missing link for understanding enzyme catalysis?". Proteins. 78 (6): 1339–75. doi:10.1002/prot.22654. PMC 2841229. PMID 20099310.
  16. ^ a b c Cermakova K, Hodges HC (May 2023). "Interaction modules that impart specificity to disordered protein". Trends in Biochemical Sciences. 48 (5): 477–490. doi:10.1016/j.tibs.2023.01.004. PMC 10106370. PMID 36754681.
  17. ^ Collins MO, Yu L, Campuzano I, Grant SG, Choudhary JS (July 2008). "Phosphoproteomic analysis of the mouse brain cytosol reveals a predominance of protein phosphorylation in regions of intrinsic sequence disorder" (PDF). Molecular & Cellular Proteomics. 7 (7): 1331–48. doi:10.1074/mcp.M700564-MCP200. PMID 18388127. S2CID 22193414.
  18. ^ Iakoucheva LM, Brown CJ, Lawson JD, Obradović Z, Dunker AK (October 2002). "Intrinsic disorder in cell-signaling and cancer-associated proteins". Journal of Molecular Biology. 323 (3): 573–84. CiteSeerX 10.1.1.132.682. doi:10.1016/S0022-2836(02)00969-5. PMID 12381310.
  19. ^ Sandhu KS (2009). "Intrinsic disorder explains diverse nuclear roles of chromatin remodeling proteins". Journal of Molecular Recognition. 22 (1): 1–8. doi:10.1002/jmr.915. PMID 18802931. S2CID 33010897.
  20. ^ Wilson BA, Foy SG, Neme R, Masel J (June 2017). "Young Genes are Highly Disordered as Predicted by the Preadaptation Hypothesis of De Novo Gene Birth". Nature Ecology & Evolution. 1 (6): 0146–146. doi:10.1038/s41559-017-0146. PMC 5476217. PMID 28642936.
  21. ^ Willis S, Masel J (September 2018). "Gene Birth Contributes to Structural Disorder Encoded by Overlapping Genes". Genetics. 210 (1): 303–313. doi:10.1534/genetics.118.301249. PMC 6116962. PMID 30026186.
  22. ^ Prakash A, Shin J, Rajan S, Yoon HS (April 2016). "Structural basis of nucleic acid recognition by FK506-binding protein 25 (FKBP25), a nuclear immunophilin". Nucleic Acids Research. 44 (6): 2909–2925. doi:10.1093/nar/gkw001. PMC 4824100. PMID 26762975.
  23. ^ Lee SH, Kim DH, Han JJ, Cha EJ, Lim JE, Cho YJ, Lee C, Han KH (February 2012). "Understanding pre-structured motifs (PresMos) in intrinsically unfolded proteins". Current Protein & Peptide Science. 13 (1): 34–54. doi:10.2174/138920312799277974. PMID 22044148.
  24. ^ Mohan A, Oldfield CJ, Radivojac P, Vacic V, Cortese MS, Dunker AK, Uversky VN (October 2006). "Analysis of molecular recognition features (MoRFs)". Journal of Molecular Biology. 362 (5): 1043–59. doi:10.1016/j.jmb.2006.07.087. PMID 16935303.
  25. ^ Gunasekaran K, Tsai CJ, Kumar S, Zanuy D, Nussinov R (February 2003). "Extended disordered proteins: targeting function with less scaffold". Trends in Biochemical Sciences. 28 (2): 81–5. doi:10.1016/S0968-0004(03)00003-3. PMID 12575995.
  26. ^ Sandhu KS, Dash D (July 2007). "Dynamic alpha-helices: conformations that do not conform". Proteins. 68 (1): 109–22. doi:10.1002/prot.21328. PMID 17407165. S2CID 96719019.
  27. ^ Tarakhovsky A, Prinjha RK (July 2018). "Drawing on disorder: How viruses use histone mimicry to their advantage". The Journal of Experimental Medicine. 215 (7): 1777–1787. doi:10.1084/jem.20180099. PMC 6028506. PMID 29934321.
  28. ^ Atkinson SC, Audsley MD, Lieu KG, Marsh GA, Thomas DR, Heaton SM, Paxman JJ, Wagstaff KM, Buckle AM, Moseley GW, Jans DA, Borg NA (January 2018). "Recognition by host nuclear transport proteins drives disorder-to-order transition in Hendra virus V". Scientific Reports. 8 (1): 358. Bibcode:2018NatSR...8..358A. doi:10.1038/s41598-017-18742-8. PMC 5762688. PMID 29321677.
  29. ^ Fuxreiter M (January 2012). "Fuzziness: linking regulation to protein dynamics". Molecular BioSystems. 8 (1): 168–77. doi:10.1039/c1mb05234a. PMID 21927770.
  30. ^ Fuxreiter M, Simon I, Bondos S (August 2011). "Dynamic protein-DNA recognition: beyond what can be seen". Trends in Biochemical Sciences. 36 (8): 415–23. doi:10.1016/j.tibs.2011.04.006. PMID 21620710.
  31. ^ Borgia A, Borgia MB, Bugge K, Kissling VM, Heidarsson PO, Fernandes CB, Sottini A, Soranno A, Buholzer KJ, Nettels D, Kragelund BB, Best RB, Schuler B (March 2018). "Extreme disorder in an ultrahigh-affinity protein complex". Nature. 555 (7694): 61–66. Bibcode:2018Natur.555...61B. doi:10.1038/nature25762. PMC 6264893. PMID 29466338.
  32. ^ Feng H, Zhou BR, Bai Y (November 2018). "Binding Affinity and Function of the Extremely Disordered Protein Complex Containing Human Linker Histone H1.0 and Its Chaperone ProTα". Biochemistry. 57 (48): 6645–6648. doi:10.1021/acs.biochem.8b01075. PMC 7984725. PMID 30430826.
  33. ^ a b Uversky VN (August 2011). "Intrinsically disordered proteins from A to Z". The International Journal of Biochemistry & Cell Biology. 43 (8): 1090–1103. doi:10.1016/j.biocel.2011.04.001. PMID 21501695.
  34. ^ a b c d Oldfield CJ, Dunker AK (2014). "Intrinsically disordered proteins and intrinsically disordered protein regions". Annual Review of Biochemistry. 83: 553–584. doi:10.1146/annurev-biochem-072711-164947. PMID 24606139.
  35. ^ Scalvini B. et al., Circuit Topology Approach for the Comparative Analysis of Intrinsically Disordered Proteins. J. Chem. Inf. Model. 63, 8, 2586–2602 (2023)
  36. ^ Theillet FX, Binolfi A, Bekei B, Martorana A, Rose HM, Stuiver M, Verzini S, Lorenz D, van Rossum M, Goldfarb D, Selenko P (2016). "Structural disorder of monomeric α-synuclein persists in mammalian cells". Nature. 530 (7588): 45–50. Bibcode:2016Natur.530...45T. doi:10.1038/nature16531. PMID 26808899. S2CID 4461465.
  37. ^ Minde DP, Ramakrishna M, Lilley KS (2018). "Biotinylation by proximity labelling favours unfolded proteins". bioRxiv. doi:10.1101/274761.
  38. ^ Minde DP, Ramakrishna M, Lilley KS (2020). "Biotin proximity tagging favours unfolded proteins and enables the study of intrinsically disordered regions". Communications Biology. 3 (1): 38. doi:10.1038/s42003-020-0758-y. PMC 6976632. PMID 31969649.
  39. ^ Minde DP, Maurice MM, Rüdiger SG (2012). Uversky VN (ed.). "Determining biophysical protein stability in lysates by a fast proteolysis assay, FASTpp". PLOS ONE. 7 (10): e46147. Bibcode:2012PLoSO...746147M. doi:10.1371/journal.pone.0046147. PMC 3463568. PMID 23056252.
  40. ^ Park C, Marqusee S (March 2005). "Pulse proteolysis: a simple method for quantitative determination of protein stability and ligand binding". Nature Methods. 2 (3): 207–12. doi:10.1038/nmeth740. PMID 15782190. S2CID 21364478.
  41. ^ Robaszkiewicz K, Ostrowska Z, Cyranka-Czaja A, Moraczewska J (May 2015). "Impaired tropomyosin-troponin interactions reduce activation of the actin thin filament". Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics. 1854 (5): 381–90. doi:10.1016/j.bbapap.2015.01.004. PMID 25603119.
  42. ^ Minde DP, Radli M, Forneris F, Maurice MM, Rüdiger SG (2013). Buckle AM (ed.). "Large extent of disorder in Adenomatous Polyposis Coli offers a strategy to guard Wnt signalling against point mutations". PLOS ONE. 8 (10): e77257. Bibcode:2013PLoSO...877257M. doi:10.1371/journal.pone.0077257. PMC 3793970. PMID 24130866.
  43. ^ Brucale M, Schuler B, Samorì B (March 2014). "Single-molecule studies of intrinsically disordered proteins". Chemical Reviews. 114 (6): 3281–317. doi:10.1021/cr400297g. PMID 24432838.
  44. ^ Neupane K, Solanki A, Sosova I, Belov M, Woodside MT (2014). "Diverse metastable structures formed by small oligomers of α-synuclein probed by force spectroscopy". PLOS ONE. 9 (1): e86495. Bibcode:2014PLoSO...986495N. doi:10.1371/journal.pone.0086495. PMC 3901707. PMID 24475132.
  45. ^ Japrung D, Dogan J, Freedman KJ, Nadzeyka A, Bauerdick S, Albrecht T, Kim MJ, Jemth P, Edel JB (February 2013). "Single-molecule studies of intrinsically disordered proteins using solid-state nanopores". Analytical Chemistry. 85 (4): 2449–56. doi:10.1021/ac3035025. PMID 23327569.
  46. ^ Min D, Kim K, Hyeon C, Cho YH, Shin YK, Yoon TY (2013). "Mechanical unzipping and rezipping of a single SNARE complex reveals hysteresis as a force-generating mechanism". Nature Communications. 4 (4): 1705. Bibcode:2013NatCo...4.1705M. doi:10.1038/ncomms2692. PMC 3644077. PMID 23591872.
  47. ^ Miyagi A, Tsunaka Y, Uchihashi T, Mayanagi K, Hirose S, Morikawa K, Ando T (September 2008). "Visualization of intrinsically disordered regions of proteins by high-speed atomic force microscopy". ChemPhysChem. 9 (13): 1859–66. doi:10.1002/cphc.200800210. PMID 18698566.
  48. ^ Campen A, Williams RM, Brown CJ, Meng J, Uversky VN, Dunker AK (2008). "TOP-IDP-scale: a new amino acid scale measuring propensity for intrinsic disorder". Protein and Peptide Letters. 15 (9): 956–963. doi:10.2174/092986608785849164. PMC 2676888. PMID 18991772.
  49. ^ Schlessinger A, Schaefer C, Vicedo E, Schmidberger M, Punta M, Rost B (June 2011). "Protein disorder--a breakthrough invention of evolution?". Current Opinion in Structural Biology. 21 (3): 412–8. doi:10.1016/j.sbi.2011.03.014. PMID 21514145.
  50. ^ Tompa P (June 2011). "Unstructural biology coming of age". Current Opinion in Structural Biology. 21 (3): 419–425. doi:10.1016/j.sbi.2011.03.012. PMID 21514142.
  51. ^ Ferron F, Longhi S, Canard B, Karlin D (October 2006). "A practical overview of protein disorder prediction methods". Proteins. 65 (1): 1–14. doi:10.1002/prot.21075. PMID 16856179. S2CID 30231497.
  52. ^ Wise-Scira O, Dunn A, Aloglu AK, Sakallioglu IT, Coskuner O (March 2013). "Structures of the E46K mutant-type α-synuclein protein and impact of E46K mutation on the structures of the wild-type α-synuclein protein". ACS Chemical Neuroscience. 4 (3): 498–508. doi:10.1021/cn3002027. PMC 3605821. PMID 23374074.
  53. ^ Dobson CM (December 2003). "Protein folding and misfolding". Nature. 426 (6968): 884–90. Bibcode:2003Natur.426..884D. doi:10.1038/nature02261. PMID 14685248. S2CID 1036192.
  54. ^ Balatti GE, Barletta GP, Parisi G, Tosatto SC, Bellanda M, Fernandez-Alberti S (December 2021). "Intrinsically Disordered Region Modulates Ligand Binding in Glutaredoxin 1 from Trypanosoma Brucei". The Journal of Physical Chemistry B. 125 (49): 13366–13375. doi:10.1021/acs.jpcb.1c07035. PMID 34870419. S2CID 244942842.
  55. ^ Best RB, Zhu X, Shim J, Lopes PE, Mittal J, Feig M, Mackerell AD (September 2012). "Optimization of the additive CHARMM all-atom protein force field targeting improved sampling of the backbone φ, ψ and side-chain χ(1) and χ(2) dihedral angles". Journal of Chemical Theory and Computation. 8 (9): 3257–3273. doi:10.1021/ct300400x. PMC 3549273. PMID 23341755.
  56. ^ Best RB (February 2017). "Computational and theoretical advances in studies of intrinsically disordered proteins". Current Opinion in Structural Biology. 42: 147–154. doi:10.1016/j.sbi.2017.01.006. PMID 28259050.
  57. ^ Chong SH, Chatterjee P, Ham S (May 2017). "Computer Simulations of Intrinsically Disordered Proteins". Annual Review of Physical Chemistry. 68: 117–134. Bibcode:2017ARPC...68..117C. doi:10.1146/annurev-physchem-052516-050843. PMID 28226222.
  58. ^ Fox SJ, Kannan S (September 2017). "Probing the dynamics of disorder". Progress in Biophysics and Molecular Biology. 128: 57–62. doi:10.1016/j.pbiomolbio.2017.05.008. PMID 28554553.
  59. ^ Terakawa T, Takada S (September 2011). "Multiscale ensemble modeling of intrinsically disordered proteins: p53 N-terminal domain". Biophysical Journal. 101 (6): 1450–1458. Bibcode:2011BpJ...101.1450T. doi:10.1016/j.bpj.2011.08.003. PMC 3177054. PMID 21943426.
  60. ^ Fisher CK, Stultz CM (June 2011). "Constructing ensembles for intrinsically disordered proteins". Current Opinion in Structural Biology. 21 (3): 426–431. doi:10.1016/j.sbi.2011.04.001. PMC 3112268. PMID 21530234.
  61. ^ Apicella A, Marascio M, Colangelo V, Soncini M, Gautieri A, Plummer CJ (June 2017). "Molecular dynamics simulations of the intrinsically disordered protein amelogenin". Journal of Biomolecular Structure & Dynamics. 35 (8): 1813–1823. doi:10.1080/07391102.2016.1196151. hdl:11311/1004711. PMID 27366858. S2CID 205576649.
  62. ^ Zerze GH, Miller CM, Granata D, Mittal J (June 2015). "Free energy surface of an intrinsically disordered protein: comparison between temperature replica exchange molecular dynamics and bias-exchange metadynamics". Journal of Chemical Theory and Computation. 11 (6): 2776–2782. doi:10.1021/acs.jctc.5b00047. PMID 26575570.
  63. ^ Granata D, Baftizadeh F, Habchi J, Galvagnion C, De Simone A, Camilloni C, et al. (October 2015). "The inverted free energy landscape of an intrinsically disordered peptide by simulations and experiments". Scientific Reports. 5: 15449. Bibcode:2015NatSR...515449G. doi:10.1038/srep15449. PMC 4620491. PMID 26498066.
  64. ^ Iida S, Kawabata T, Kasahara K, Nakamura H, Higo J (April 2019). "Multimodal Structural Distribution of the p53 C-Terminal Domain upon Binding to S100B via a Generalized Ensemble Method: From Disorder to Extradisorder". Journal of Chemical Theory and Computation. 15 (4): 2597–2607. doi:10.1021/acs.jctc.8b01042. PMID 30855964. S2CID 75138292.
  65. ^ Kurcinski M, Kolinski A, Kmiecik S (June 2014). "Mechanism of Folding and Binding of an Intrinsically Disordered Protein As Revealed by ab Initio Simulations". Journal of Chemical Theory and Computation. 10 (6): 2224–2231. doi:10.1021/ct500287c. PMID 26580746.
  66. ^ Ciemny MP, Badaczewska-Dawid AE, Pikuzinska M, Kolinski A, Kmiecik S (January 2019). "Modeling of Disordered Protein Structures Using Monte Carlo Simulations and Knowledge-Based Statistical Force Fields". International Journal of Molecular Sciences. 20 (3): 606. doi:10.3390/ijms20030606. PMC 6386871. PMID 30708941.
  67. ^ Garaizar A, Espinosa JR (September 2021). "Salt dependent phase behavior of intrinsically disordered proteins from a coarse-grained model with explicit water and ions". The Journal of Chemical Physics. 155 (12): 125103. Bibcode:2021JChPh.155l5103G. doi:10.1063/5.0062687. PMID 34598583. S2CID 238249229.
  68. ^ Uversky VN (2013). "Digested disorder: Quarterly intrinsic disorder digest (January/February/March, 2013)". Intrinsically Disordered Proteins. 1 (1): e25496. doi:10.4161/idp.25496. PMC 5424799. PMID 28516015.
  69. ^ Costantini S, Sharma A, Raucci R, Costantini M, Autiero I, Colonna G (March 2013). "Genealogy of an ancient protein family: the Sirtuins, a family of disordered members". BMC Evolutionary Biology. 13: 60. doi:10.1186/1471-2148-13-60. PMC 3599600. PMID 23497088.

External links edit

  • Intrinsically disordered protein at Proteopedia
  • MobiDB: a comprehensive database of intrinsic protein disorder annotations
  • IDEAL - Intrinsically Disordered proteins with Extensive Annotations and Literature 2020-05-02 at the Wayback Machine
  • D2P2 Database of Disordered Protein Predictions
  • First IDP journal covering all topics of IDP research
  • IDP Journal
  • Database of experimentally validated IDPs
  • IDP ensemble database 2018-03-10 at the Wayback Machine

intrinsically, disordered, proteins, molecular, biology, intrinsically, disordered, protein, protein, that, lacks, fixed, ordered, three, dimensional, structure, typically, absence, macromolecular, interaction, partners, such, other, proteins, idps, range, fro. In molecular biology an intrinsically disordered protein IDP is a protein that lacks a fixed or ordered three dimensional structure 2 3 4 typically in the absence of its macromolecular interaction partners such as other proteins or RNA IDPs range from fully unstructured to partially structured and include random coil molten globule like aggregates or flexible linkers in large multi domain proteins They are sometimes considered as a separate class of proteins along with globular fibrous and membrane proteins 5 Conformational flexibility in SUMO 1 protein PDB 1a5r The central part shows relatively ordered structure Conversely the N and C terminal regions left and right respectively show intrinsic disorder although a short helical region persists in the N terminal tail Ten alternative NMR models were morphed Secondary structure elements a helices red b strands blue arrows 1 IDPs are a very large and functionally important class of proteins and their discovery has disproved the idea that three dimensional structures of proteins must be fixed to accomplish their biological functions For example IDPs have been identified to participate in weak multivalent interactions that are highly cooperative and dynamic lending them importance in DNA regulation and in cell signaling 6 7 Many IDPs can also adopt a fixed three dimensional structure after binding to other macromolecules Overall IDPs are different from structured proteins in many ways and tend to have distinctive function structure sequence interactions evolution and regulation 8 Contents 1 History 2 Abundance 3 Biological roles 3 1 Flexible linkers 3 2 Linear motifs 3 3 Pre structured motifs 3 4 Coupled folding and binding 3 5 Disorder in the bound state fuzzy complexes 4 Structural aspects 5 Experimental validation 5 1 In vivo approaches 5 2 In vitro approaches 6 Disorder annotation 6 1 Disorder databases 6 2 Predicting IDPs by sequence 6 3 Prediction methods 7 Disorder and disease 8 Computer simulations 9 See also 10 References 11 External linksHistory edit nbsp An ensemble of NMR structures of the Thylakoid soluble phosphoprotein TSP9 which shows a largely flexible protein chain 9 In the 1930s 1950s the first protein structures were solved by protein crystallography These early structures suggested that a fixed three dimensional structure might be generally required to mediate biological functions of proteins These publications solidified the central dogma of molecular biology in that the amino acid sequence of a protein determines its structure which in turn determines its function In 1950 Karush wrote about Configurational Adaptability contradicting this assumption He was convinced that proteins have more than one configuration at the same energy level and can choose one when binding to other substrates In the 1960s Levinthal s paradox suggested that the systematic conformational search of a long polypeptide is unlikely to yield a single folded protein structure on biologically relevant timescales i e microseconds to minutes Curiously for many small proteins or protein domains relatively rapid and efficient refolding can be observed in vitro As stated in Anfinsen s Dogma from 1973 the fixed 3D structure of these proteins is uniquely encoded in its primary structure the amino acid sequence is kinetically accessible and stable under a range of near physiological conditions and can therefore be considered as the native state of such ordered proteins 10 During the subsequent decades however many large protein regions could not be assigned in x ray datasets indicating that they occupy multiple positions which average out in electron density maps The lack of fixed unique positions relative to the crystal lattice suggested that these regions were disordered Nuclear magnetic resonance spectroscopy of proteins also demonstrated the presence of large flexible linkers and termini in many solved structural ensembles In 2001 Dunker questioned whether the newly found information was ignored for 50 years 11 with more quantitative analyses becoming available in the 2000s 12 In the 2010s it became clear that IDPs are common among disease related proteins such as alpha synuclein and tau 13 Abundance editIt is now generally accepted that proteins exist as an ensemble of similar structures with some regions more constrained than others IDPs occupy the extreme end of this spectrum of flexibility and include proteins of considerable local structure tendency or flexible multidomain assemblies 14 15 Intrinsic disorder is particularly elevated among proteins that regulate chromatin and transcription 16 and bioinformatic predictions indicate that is more common in genomes and proteomes than in known structures in the protein database Based on DISOPRED2 prediction long gt 30 residue disordered segments occur in 2 0 of archaean 4 2 of eubacterial and 33 0 of eukaryotic proteins 12 including certain disease related proteins 13 Biological roles editHighly dynamic disordered regions of proteins have been linked to functionally important phenomena such as allosteric regulation and enzyme catalysis 14 15 Many disordered proteins have the binding affinity with their receptors regulated by post translational modification thus it has been proposed that the flexibility of disordered proteins facilitates the different conformational requirements for binding the modifying enzymes as well as their receptors 17 Intrinsic disorder is particularly enriched in proteins implicated in cell signaling and transcription 16 as well as chromatin remodeling functions 18 19 Genes that have recently been born de novo tend to have higher disorder 20 21 Flexible linkers edit Disordered regions are often found as flexible linkers or loops connecting domains Linker sequences vary greatly in length but are typically rich in polar uncharged amino acids Flexible linkers allow the connecting domains to freely twist and rotate to recruit their binding partners via protein domain dynamics They also allow their binding partners to induce larger scale conformational changes by long range allostery 14 2 The flexible linker of FBP25 which connects two domains of FKBP25 is important for the binding of FKBP25 with DNA 22 Linear motifs edit Linear motifs are short disordered segments of proteins that mediate functional interactions with other proteins or other biomolecules RNA DNA sugars etc 16 Many roles of linear motifs are associated with cell regulation for instance in control of cell shape subcellular localisation of individual proteins and regulated protein turnover Often post translational modifications such as phosphorylation tune the affinity not rarely by several orders of magnitude of individual linear motifs for specific interactions Relatively rapid evolution and a relatively small number of structural restraints for establishing novel low affinity interfaces make it particularly challenging to detect linear motifs but their widespread biological roles and the fact that many viruses mimick hijack linear motifs to efficiently recode infected cells underlines the timely urgency of research on this very challenging and exciting topic Pre structured motifs edit Unlike globular proteins IDPs do not have spatially disposed active pockets Fascinatingly 80 of target unbound IDPs 4 dozens subjected to detailed structural characterization by NMR possess linear motifs termed PresMos pre structured motifs 23 that are transient secondary structural elements primed for target recognition In several cases it has been demonstrated that these transient structures become full and stable secondary structures e g helices upon target binding Hence PresMos are the putative active sites in IDPs Coupled folding and binding edit Many unstructured proteins undergo transitions to more ordered states upon binding to their targets e g Molecular Recognition Features MoRFs 24 The coupled folding and binding may be local involving only a few interacting residues or it might involve an entire protein domain It was recently shown that the coupled folding and binding allows the burial of a large surface area that would be possible only for fully structured proteins if they were much larger 25 Moreover certain disordered regions might serve as molecular switches in regulating certain biological function by switching to ordered conformation upon molecular recognition like small molecule binding DNA RNA binding ion interactions etc 26 The ability of disordered proteins to bind and thus to exert a function shows that stability is not a required condition Many short functional sites for example Short Linear Motifs are over represented in disordered proteins Disordered proteins and short linear motifs are particularly abundant in many RNA viruses such as Hendra virus HCV HIV 1 and human papillomaviruses This enables such viruses to overcome their informationally limited genomes by facilitating binding and manipulation of a large number of host cell proteins 27 28 Disorder in the bound state fuzzy complexes edit Intrinsically disordered proteins can retain their conformational freedom even when they bind specifically to other proteins The structural disorder in bound state can be static or dynamic In fuzzy complexes structural multiplicity is required for function and the manipulation of the bound disordered region changes activity The conformational ensemble of the complex is modulated via post translational modifications or protein interactions 29 Specificity of DNA binding proteins often depends on the length of fuzzy regions which is varied by alternative splicing 30 Some fuzzy complexes may exhibit high binding affinity 31 although other studies showed different affinity values for the same system in a different concentration regime 32 Structural aspects editIntrinsically disordered proteins adapt many different structures in vivo according to the cell s conditions creating a structural or conformational ensemble 33 34 Therefore their structures are strongly function related However only few proteins are fully disordered in their native state Disorder is mostly found in intrinsically disordered regions IDRs within an otherwise well structured protein The term intrinsically disordered protein IDP therefore includes proteins that contain IDRs as well as fully disordered proteins The existence and kind of protein disorder is encoded in its amino acid sequence 2 In general IDPs are characterized by a low content of bulky hydrophobic amino acids and a high proportion of polar and charged amino acids usually referred to as low hydrophobicity 33 This property leads to good interactions with water Furthermore high net charges promote disorder because of electrostatic repulsion resulting from equally charged residues 34 Thus disordered sequences cannot sufficiently bury a hydrophobic core to fold into stable globular proteins In some cases hydrophobic clusters in disordered sequences provide the clues for identifying the regions that undergo coupled folding and binding refer to biological roles Many disordered proteins reveal regions without any regular secondary structure These regions can be termed as flexible compared to structured loops While the latter are rigid and contain only one set of Ramachandran angles IDPs involve multiple sets of angles 34 The term flexibility is also used for well structured proteins but describes a different phenomenon in the context of disordered proteins Flexibility in structured proteins is bound to an equilibrium state while it is not so in IDPs 34 Many disordered proteins also reveal low complexity sequences i e sequences with over representation of a few residues While low complexity sequences are a strong indication of disorder the reverse is not necessarily true that is not all disordered proteins have low complexity sequences Disordered proteins have a low content of predicted secondary structure Due to the disordered nature of these proteins topological approaches have been developed to search for conformational patterns in their dynamics For instance circuit topology has been applied to track the dynamics of disordered protein domains 35 By employing a topological approach one can categorize motifs according to their topological buildup and the timescale of their formation Experimental validation editIDPs can be validated in several contexts Most approaches for experimental validation of IDPs are restricted to extracted or purified proteins while some new experimental strategies aim to explore in vivo conformations and structural variations of IDPs inside intact living cells and systematic comparisons between their dynamics in vivo and in vitro In vivo approaches edit The first direct evidence for in vivo persistence of intrinsic disorder has been achieved by in cell NMR upon electroporation of a purified IDP and recovery of cells to an intact state 36 Larger scale in vivo validation of IDR predictions is now possible using biotin painting 37 38 In vitro approaches edit Intrinsically unfolded proteins once purified can be identified by various experimental methods The primary method to obtain information on disordered regions of a protein is NMR spectroscopy The lack of electron density in X ray crystallographic studies may also be a sign of disorder Folded proteins have a high density partial specific volume of 0 72 0 74 mL g and commensurately small radius of gyration Hence unfolded proteins can be detected by methods that are sensitive to molecular size density or hydrodynamic drag such as size exclusion chromatography analytical ultracentrifugation small angle X ray scattering SAXS and measurements of the diffusion constant Unfolded proteins are also characterized by their lack of secondary structure as assessed by far UV 170 250 nm circular dichroism esp a pronounced minimum at 200 nm or infrared spectroscopy Unfolded proteins also have exposed backbone peptide groups exposed to solvent so that they are readily cleaved by proteases undergo rapid hydrogen deuterium exchange and exhibit a small dispersion lt 1 ppm in their 1H amide chemical shifts as measured by NMR Folded proteins typically show dispersions as large as 5 ppm for the amide protons Recently new methods including Fast parallel proteolysis FASTpp have been introduced which allow to determine the fraction folded disordered without the need for purification 39 40 Even subtle differences in the stability of missense mutations protein partner binding and self polymerisation induced folding of e g coiled coils can be detected using FASTpp as recently demonstrated using the tropomyosin troponin protein interaction 41 Fully unstructured protein regions can be experimentally validated by their hypersusceptibility to proteolysis using short digestion times and low protease concentrations 42 Bulk methods to study IDP structure and dynamics include SAXS for ensemble shape information NMR for atomistic ensemble refinement Fluorescence for visualising molecular interactions and conformational transitions x ray crystallography to highlight more mobile regions in otherwise rigid protein crystals cryo EM to reveal less fixed parts of proteins light scattering to monitor size distributions of IDPs or their aggregation kinetics NMR chemical shift and Circular Dichroism to monitor secondary structure of IDPs Single molecule methods to study IDPs include spFRET 43 to study conformational flexibility of IDPs and the kinetics of structural transitions optical tweezers 44 for high resolution insights into the ensembles of IDPs and their oligomers or aggregates nanopores 45 to reveal global shape distributions of IDPs magnetic tweezers 46 to study structural transitions for long times at low forces high speed AFM 47 to visualise the spatio temporal flexibility of IDPs directly Disorder annotation edit nbsp REMARK465 missing electron densities in X ray structure representing protein disorder PDB 1a22 human growth hormone bound to receptor Compilation of screenshots from PDB database and molecule representation via VMD Blue and red arrows point to missing residues on receptor and growth hormone respectively Intrinsic disorder can be either annotated from experimental information or predicted with specialized software Disorder prediction algorithms can predict Intrinsic Disorder ID propensity with high accuracy approaching around 80 based on primary sequence composition similarity to unassigned segments in protein x ray datasets flexible regions in NMR studies and physico chemical properties of amino acids Disorder databases edit Databases have been established to annotate protein sequences with intrinsic disorder information The DisProt database contains a collection of manually curated protein segments which have been experimentally determined to be disordered MobiDB is a database combining experimentally curated disorder annotations e g from DisProt with data derived from missing residues in X ray crystallographic structures and flexible regions in NMR structures Predicting IDPs by sequence edit Separating disordered from ordered proteins is essential for disorder prediction One of the first steps to find a factor that distinguishes IDPs from non IDPs is to specify biases within the amino acid composition The following hydrophilic charged amino acids A R G Q S P E and K have been characterized as disorder promoting amino acids while order promoting amino acids W C F I Y V L and N are hydrophobic and uncharged The remaining amino acids H M T and D are ambiguous found in both ordered and unstructured regions 2 A more recent analysis ranked amino acids by their propensity to form disordered regions as follows order promoting to disorder promoting W F Y I M L V N C T A G R D H Q K S E P 48 As it can be seen from the list small charged hydrophilic residues often promote disorder while large and hydrophobic residues promote order This information is the basis of most sequence based predictors Regions with little to no secondary structure also known as NORS NO Regular Secondary structure regions 49 and low complexity regions can easily be detected However not all disordered proteins contain such low complexity sequences Prediction methods edit Main article List of disorder prediction software Determining disordered regions from biochemical methods is very costly and time consuming Due to the variable nature of IDPs only certain aspects of their structure can be detected so that a full characterization requires a large number of different methods and experiments This further increases the expense of IDP determination In order to overcome this obstacle computer based methods are created for predicting protein structure and function It is one of the main goals of bioinformatics to derive knowledge by prediction Predictors for IDP function are also being developed but mainly use structural information such as linear motif sites 4 50 There are different approaches for predicting IDP structure such as neural networks or matrix calculations based on different structural and or biophysical properties Many computational methods exploit sequence information to predict whether a protein is disordered 51 Notable examples of such software include IUPRED and Disopred Different methods may use different definitions of disorder Meta predictors show a new concept combining different primary predictors to create a more competent and exact predictor Due to the different approaches of predicting disordered proteins estimating their relative accuracy is fairly difficult For example neural networks are often trained on different datasets The disorder prediction category is a part of biannual CASP experiment that is designed to test methods according accuracy in finding regions with missing 3D structure marked in PDB files as REMARK465 missing electron densities in X ray structures Disorder and disease editIntrinsically unstructured proteins have been implicated in a number of diseases 13 Aggregation of misfolded proteins is the cause of many synucleinopathies and toxicity as those proteins start binding to each other randomly and can lead to cancer or cardiovascular diseases Thereby misfolding can happen spontaneously because millions of copies of proteins are made during the lifetime of an organism The aggregation of the intrinsically unstructured protein a synuclein is thought to be responsible The structural flexibility of this protein together with its susceptibility to modification in the cell leads to misfolding and aggregation Genetics oxidative and nitrative stress as well as mitochondrial impairment impact the structural flexibility of the unstructured a synuclein protein and associated disease mechanisms 52 Many key tumour suppressors have large intrinsically unstructured regions for example p53 and BRCA1 These regions of the proteins are responsible for mediating many of their interactions Taking the cell s native defense mechanisms as a model drugs can be developed trying to block the place of noxious substrates and inhibiting them and thus counteracting the disease 53 Computer simulations edit nbsp MD simulation of the Glutaredoxin 1 from Trypanosoma Brucei The globular thioredoxin fold is depicted in blue while the disordered N tail in green According to the MD results the disordered tail can be modulating the dynamics of the binding pocket 54 Owing to high structural heterogeneity NMR SAXS experimental parameters obtained will be an average over a large number of highly diverse and disordered states an ensemble of disordered states Hence to understand the structural implications of these experimental parameters there is a necessity for accurate representation of these ensembles by computer simulations All atom molecular dynamic simulations can be used for this purpose but their use is limited by the accuracy of current force fields in representing disordered proteins Nevertheless some force fields have been explicitly developed for studying disordered proteins by optimising force field parameters using available NMR data for disordered proteins examples are CHARMM 22 CHARMM 32 55 Amber ff03 etc MD simulations restrained by experimental parameters restrained MD have also been used to characterise disordered proteins 56 57 58 In principle one can sample the whole conformational space given an MD simulation with accurate Force field is run long enough Because of very high structural heterogeneity the time scales that needs to be run for this purpose are very large and are limited by computational power However other computational techniques such as accelerated MD simulations 59 replica exchange simulations 60 61 metadynamics 62 63 multicanonical MD simulations 64 or methods using coarse grained representation with implicit and explicit solvents 65 66 67 have been used to sample broader conformational space in smaller time scales Moreover various protocols and methods of analyzing IDPs such as studies based on quantitative analysis of GC content in genes and their respective chromosomal bands have been used to understand functional IDP segments 68 69 See also editIDPbyNMR DisProt database MobiDB database Molten globule Random coil Dark proteomeReferences edit Majorek K Kozlowski L Jakalski M Bujnicki JM December 18 2008 First Steps of Protein Structure Prediction PDF In Bujnicki J ed Prediction of Protein Structures Functions and Interactions John Wiley amp Sons Ltd pp 39 62 doi 10 1002 9780470741894 ch2 ISBN 9780470517673 a b c d Dunker AK Lawson JD Brown CJ Williams RM Romero P Oh JS Oldfield CJ Campen AM Ratliff CM Hipps KW Ausio J Nissen MS Reeves R Kang C Kissinger CR Bailey RW Griswold MD Chiu W Garner EC Obradovic Z 2001 Intrinsically disordered protein Journal of Molecular Graphics amp Modelling 19 1 26 59 CiteSeerX 10 1 1 113 556 doi 10 1016 s1093 3263 00 00138 8 PMID 11381529 Dyson HJ Wright PE March 2005 Intrinsically unstructured proteins and their functions Nature Reviews Molecular Cell Biology 6 3 197 208 doi 10 1038 nrm1589 PMID 15738986 S2CID 18068406 a b Dunker AK Silman I Uversky VN Sussman JL December 2008 Function and structure of inherently disordered proteins Current Opinion in Structural Biology 18 6 756 64 doi 10 1016 j sbi 2008 10 002 PMID 18952168 Andreeva A Howorth D Chothia C Kulesha E Murzin AG January 2014 SCOP2 prototype a new approach to protein structure mining Nucleic Acids Research 42 Database issue D310 4 doi 10 1093 nar gkt1242 PMC 3964979 PMID 24293656 Mir M Stadler MR Ortiz SA Hannon CE Harrison MM Darzacq X Eisen MB December 2018 Singer RH Struhl K Crocker J eds Dynamic multifactor hubs interact transiently with sites of active transcription in Drosophila embryos eLife 7 e40497 doi 10 7554 eLife 40497 PMC 6307861 PMID 30589412 Wright PE Dyson HJ January 2015 Intrinsically disordered proteins in cellular signalling and regulation Nature Reviews Molecular Cell Biology 16 1 18 29 doi 10 1038 nrm3920 PMC 4405151 PMID 25531225 van der Lee R Buljan M Lang B Weatheritt RJ Daughdrill GW Dunker AK et al July 2014 Classification of intrinsically disordered regions and proteins Chemical Reviews 114 13 6589 6631 doi 10 1021 cr400525m PMC 4095912 PMID 24773235 Song J Lee MS Carlberg I Vener AV Markley JL December 2006 Micelle induced folding of spinach thylakoid soluble phosphoprotein of 9 kDa and its functional implications Biochemistry 45 51 15633 43 doi 10 1021 bi062148m PMC 2533273 PMID 17176085 Anfinsen CB July 1973 Principles that govern the folding of protein chains Science 181 4096 223 230 Bibcode 1973Sci 181 223A doi 10 1126 science 181 4096 223 PMID 4124164 Dunker AK Lawson JD Brown CJ Williams RM Romero P Oh JS Oldfield CJ Campen AM Ratliff CM Hipps KW Ausio J Nissen MS Reeves R Kang C Kissinger CR Bailey RW Griswold MD Chiu W Garner EC Obradovic Z 2001 01 01 Intrinsically disordered protein Journal of Molecular Graphics amp Modelling 19 1 26 59 CiteSeerX 10 1 1 113 556 doi 10 1016 s1093 3263 00 00138 8 PMID 11381529 a b Ward JJ Sodhi JS McGuffin LJ Buxton BF Jones DT March 2004 Prediction and functional analysis of native disorder in proteins from the three kingdoms of life Journal of Molecular Biology 337 3 635 45 CiteSeerX 10 1 1 120 5605 doi 10 1016 j jmb 2004 02 002 PMID 15019783 a b c Uversky VN Oldfield CJ Dunker AK 2008 Intrinsically disordered proteins in human diseases introducing the D2 concept Annual Review of Biophysics 37 215 46 doi 10 1146 annurev biophys 37 032807 125924 PMID 18573080 a b c Bu Z Callaway DJ 2011 Proteins MOVE Protein dynamics and long range allostery in cell signaling Protein Structure and Diseases Advances in Protein Chemistry and Structural Biology Vol 83 pp 163 221 doi 10 1016 B978 0 12 381262 9 00005 7 ISBN 9780123812629 PMID 21570668 a b Kamerlin SC Warshel A May 2010 At the dawn of the 21st century Is dynamics the missing link for understanding enzyme catalysis Proteins 78 6 1339 75 doi 10 1002 prot 22654 PMC 2841229 PMID 20099310 a b c Cermakova K Hodges HC May 2023 Interaction modules that impart specificity to disordered protein Trends in Biochemical Sciences 48 5 477 490 doi 10 1016 j tibs 2023 01 004 PMC 10106370 PMID 36754681 Collins MO Yu L Campuzano I Grant SG Choudhary JS July 2008 Phosphoproteomic analysis of the mouse brain cytosol reveals a predominance of protein phosphorylation in regions of intrinsic sequence disorder PDF Molecular amp Cellular Proteomics 7 7 1331 48 doi 10 1074 mcp M700564 MCP200 PMID 18388127 S2CID 22193414 Iakoucheva LM Brown CJ Lawson JD Obradovic Z Dunker AK October 2002 Intrinsic disorder in cell signaling and cancer associated proteins Journal of Molecular Biology 323 3 573 84 CiteSeerX 10 1 1 132 682 doi 10 1016 S0022 2836 02 00969 5 PMID 12381310 Sandhu KS 2009 Intrinsic disorder explains diverse nuclear roles of chromatin remodeling proteins Journal of Molecular Recognition 22 1 1 8 doi 10 1002 jmr 915 PMID 18802931 S2CID 33010897 Wilson BA Foy SG Neme R Masel J June 2017 Young Genes are Highly Disordered as Predicted by the Preadaptation Hypothesis of De Novo Gene Birth Nature Ecology amp Evolution 1 6 0146 146 doi 10 1038 s41559 017 0146 PMC 5476217 PMID 28642936 Willis S Masel J September 2018 Gene Birth Contributes to Structural Disorder Encoded by Overlapping Genes Genetics 210 1 303 313 doi 10 1534 genetics 118 301249 PMC 6116962 PMID 30026186 Prakash A Shin J Rajan S Yoon HS April 2016 Structural basis of nucleic acid recognition by FK506 binding protein 25 FKBP25 a nuclear immunophilin Nucleic Acids Research 44 6 2909 2925 doi 10 1093 nar gkw001 PMC 4824100 PMID 26762975 Lee SH Kim DH Han JJ Cha EJ Lim JE Cho YJ Lee C Han KH February 2012 Understanding pre structured motifs PresMos in intrinsically unfolded proteins Current Protein amp Peptide Science 13 1 34 54 doi 10 2174 138920312799277974 PMID 22044148 Mohan A Oldfield CJ Radivojac P Vacic V Cortese MS Dunker AK Uversky VN October 2006 Analysis of molecular recognition features MoRFs Journal of Molecular Biology 362 5 1043 59 doi 10 1016 j jmb 2006 07 087 PMID 16935303 Gunasekaran K Tsai CJ Kumar S Zanuy D Nussinov R February 2003 Extended disordered proteins targeting function with less scaffold Trends in Biochemical Sciences 28 2 81 5 doi 10 1016 S0968 0004 03 00003 3 PMID 12575995 Sandhu KS Dash D July 2007 Dynamic alpha helices conformations that do not conform Proteins 68 1 109 22 doi 10 1002 prot 21328 PMID 17407165 S2CID 96719019 Tarakhovsky A Prinjha RK July 2018 Drawing on disorder How viruses use histone mimicry to their advantage The Journal of Experimental Medicine 215 7 1777 1787 doi 10 1084 jem 20180099 PMC 6028506 PMID 29934321 Atkinson SC Audsley MD Lieu KG Marsh GA Thomas DR Heaton SM Paxman JJ Wagstaff KM Buckle AM Moseley GW Jans DA Borg NA January 2018 Recognition by host nuclear transport proteins drives disorder to order transition in Hendra virus V Scientific Reports 8 1 358 Bibcode 2018NatSR 8 358A doi 10 1038 s41598 017 18742 8 PMC 5762688 PMID 29321677 Fuxreiter M January 2012 Fuzziness linking regulation to protein dynamics Molecular BioSystems 8 1 168 77 doi 10 1039 c1mb05234a PMID 21927770 Fuxreiter M Simon I Bondos S August 2011 Dynamic protein DNA recognition beyond what can be seen Trends in Biochemical Sciences 36 8 415 23 doi 10 1016 j tibs 2011 04 006 PMID 21620710 Borgia A Borgia MB Bugge K Kissling VM Heidarsson PO Fernandes CB Sottini A Soranno A Buholzer KJ Nettels D Kragelund BB Best RB Schuler B March 2018 Extreme disorder in an ultrahigh affinity protein complex Nature 555 7694 61 66 Bibcode 2018Natur 555 61B doi 10 1038 nature25762 PMC 6264893 PMID 29466338 Feng H Zhou BR Bai Y November 2018 Binding Affinity and Function of the Extremely Disordered Protein Complex Containing Human Linker Histone H1 0 and Its Chaperone ProTa Biochemistry 57 48 6645 6648 doi 10 1021 acs biochem 8b01075 PMC 7984725 PMID 30430826 a b Uversky VN August 2011 Intrinsically disordered proteins from A to Z The International Journal of Biochemistry amp Cell Biology 43 8 1090 1103 doi 10 1016 j biocel 2011 04 001 PMID 21501695 a b c d Oldfield CJ Dunker AK 2014 Intrinsically disordered proteins and intrinsically disordered protein regions Annual Review of Biochemistry 83 553 584 doi 10 1146 annurev biochem 072711 164947 PMID 24606139 Scalvini B et al Circuit Topology Approach for the Comparative Analysis of Intrinsically Disordered Proteins J Chem Inf Model 63 8 2586 2602 2023 Theillet FX Binolfi A Bekei B Martorana A Rose HM Stuiver M Verzini S Lorenz D van Rossum M Goldfarb D Selenko P 2016 Structural disorder of monomeric a synuclein persists in mammalian cells Nature 530 7588 45 50 Bibcode 2016Natur 530 45T doi 10 1038 nature16531 PMID 26808899 S2CID 4461465 Minde DP Ramakrishna M Lilley KS 2018 Biotinylation by proximity labelling favours unfolded proteins bioRxiv doi 10 1101 274761 Minde DP Ramakrishna M Lilley KS 2020 Biotin proximity tagging favours unfolded proteins and enables the study of intrinsically disordered regions Communications Biology 3 1 38 doi 10 1038 s42003 020 0758 y PMC 6976632 PMID 31969649 Minde DP Maurice MM Rudiger SG 2012 Uversky VN ed Determining biophysical protein stability in lysates by a fast proteolysis assay FASTpp PLOS ONE 7 10 e46147 Bibcode 2012PLoSO 746147M doi 10 1371 journal pone 0046147 PMC 3463568 PMID 23056252 Park C Marqusee S March 2005 Pulse proteolysis a simple method for quantitative determination of protein stability and ligand binding Nature Methods 2 3 207 12 doi 10 1038 nmeth740 PMID 15782190 S2CID 21364478 Robaszkiewicz K Ostrowska Z Cyranka Czaja A Moraczewska J May 2015 Impaired tropomyosin troponin interactions reduce activation of the actin thin filament Biochimica et Biophysica Acta BBA Proteins and Proteomics 1854 5 381 90 doi 10 1016 j bbapap 2015 01 004 PMID 25603119 Minde DP Radli M Forneris F Maurice MM Rudiger SG 2013 Buckle AM ed Large extent of disorder in Adenomatous Polyposis Coli offers a strategy to guard Wnt signalling against point mutations PLOS ONE 8 10 e77257 Bibcode 2013PLoSO 877257M doi 10 1371 journal pone 0077257 PMC 3793970 PMID 24130866 Brucale M Schuler B Samori B March 2014 Single molecule studies of intrinsically disordered proteins Chemical Reviews 114 6 3281 317 doi 10 1021 cr400297g PMID 24432838 Neupane K Solanki A Sosova I Belov M Woodside MT 2014 Diverse metastable structures formed by small oligomers of a synuclein probed by force spectroscopy PLOS ONE 9 1 e86495 Bibcode 2014PLoSO 986495N doi 10 1371 journal pone 0086495 PMC 3901707 PMID 24475132 Japrung D Dogan J Freedman KJ Nadzeyka A Bauerdick S Albrecht T Kim MJ Jemth P Edel JB February 2013 Single molecule studies of intrinsically disordered proteins using solid state nanopores Analytical Chemistry 85 4 2449 56 doi 10 1021 ac3035025 PMID 23327569 Min D Kim K Hyeon C Cho YH Shin YK Yoon TY 2013 Mechanical unzipping and rezipping of a single SNARE complex reveals hysteresis as a force generating mechanism Nature Communications 4 4 1705 Bibcode 2013NatCo 4 1705M doi 10 1038 ncomms2692 PMC 3644077 PMID 23591872 Miyagi A Tsunaka Y Uchihashi T Mayanagi K Hirose S Morikawa K Ando T September 2008 Visualization of intrinsically disordered regions of proteins by high speed atomic force microscopy ChemPhysChem 9 13 1859 66 doi 10 1002 cphc 200800210 PMID 18698566 Campen A Williams RM Brown CJ Meng J Uversky VN Dunker AK 2008 TOP IDP scale a new amino acid scale measuring propensity for intrinsic disorder Protein and Peptide Letters 15 9 956 963 doi 10 2174 092986608785849164 PMC 2676888 PMID 18991772 Schlessinger A Schaefer C Vicedo E Schmidberger M Punta M Rost B June 2011 Protein disorder a breakthrough invention of evolution Current Opinion in Structural Biology 21 3 412 8 doi 10 1016 j sbi 2011 03 014 PMID 21514145 Tompa P June 2011 Unstructural biology coming of age Current Opinion in Structural Biology 21 3 419 425 doi 10 1016 j sbi 2011 03 012 PMID 21514142 Ferron F Longhi S Canard B Karlin D October 2006 A practical overview of protein disorder prediction methods Proteins 65 1 1 14 doi 10 1002 prot 21075 PMID 16856179 S2CID 30231497 Wise Scira O Dunn A Aloglu AK Sakallioglu IT Coskuner O March 2013 Structures of the E46K mutant type a synuclein protein and impact of E46K mutation on the structures of the wild type a synuclein protein ACS Chemical Neuroscience 4 3 498 508 doi 10 1021 cn3002027 PMC 3605821 PMID 23374074 Dobson CM December 2003 Protein folding and misfolding Nature 426 6968 884 90 Bibcode 2003Natur 426 884D doi 10 1038 nature02261 PMID 14685248 S2CID 1036192 Balatti GE Barletta GP Parisi G Tosatto SC Bellanda M Fernandez Alberti S December 2021 Intrinsically Disordered Region Modulates Ligand Binding in Glutaredoxin 1 from Trypanosoma Brucei The Journal of Physical Chemistry B 125 49 13366 13375 doi 10 1021 acs jpcb 1c07035 PMID 34870419 S2CID 244942842 Best RB Zhu X Shim J Lopes PE Mittal J Feig M Mackerell AD September 2012 Optimization of the additive CHARMM all atom protein force field targeting improved sampling of the backbone f ps and side chain x 1 and x 2 dihedral angles Journal of Chemical Theory and Computation 8 9 3257 3273 doi 10 1021 ct300400x PMC 3549273 PMID 23341755 Best RB February 2017 Computational and theoretical advances in studies of intrinsically disordered proteins Current Opinion in Structural Biology 42 147 154 doi 10 1016 j sbi 2017 01 006 PMID 28259050 Chong SH Chatterjee P Ham S May 2017 Computer Simulations of Intrinsically Disordered Proteins Annual Review of Physical Chemistry 68 117 134 Bibcode 2017ARPC 68 117C doi 10 1146 annurev physchem 052516 050843 PMID 28226222 Fox SJ Kannan S September 2017 Probing the dynamics of disorder Progress in Biophysics and Molecular Biology 128 57 62 doi 10 1016 j pbiomolbio 2017 05 008 PMID 28554553 Terakawa T Takada S September 2011 Multiscale ensemble modeling of intrinsically disordered proteins p53 N terminal domain Biophysical Journal 101 6 1450 1458 Bibcode 2011BpJ 101 1450T doi 10 1016 j bpj 2011 08 003 PMC 3177054 PMID 21943426 Fisher CK Stultz CM June 2011 Constructing ensembles for intrinsically disordered proteins Current Opinion in Structural Biology 21 3 426 431 doi 10 1016 j sbi 2011 04 001 PMC 3112268 PMID 21530234 Apicella A Marascio M Colangelo V Soncini M Gautieri A Plummer CJ June 2017 Molecular dynamics simulations of the intrinsically disordered protein amelogenin Journal of Biomolecular Structure amp Dynamics 35 8 1813 1823 doi 10 1080 07391102 2016 1196151 hdl 11311 1004711 PMID 27366858 S2CID 205576649 Zerze GH Miller CM Granata D Mittal J June 2015 Free energy surface of an intrinsically disordered protein comparison between temperature replica exchange molecular dynamics and bias exchange metadynamics Journal of Chemical Theory and Computation 11 6 2776 2782 doi 10 1021 acs jctc 5b00047 PMID 26575570 Granata D Baftizadeh F Habchi J Galvagnion C De Simone A Camilloni C et al October 2015 The inverted free energy landscape of an intrinsically disordered peptide by simulations and experiments Scientific Reports 5 15449 Bibcode 2015NatSR 515449G doi 10 1038 srep15449 PMC 4620491 PMID 26498066 Iida S Kawabata T Kasahara K Nakamura H Higo J April 2019 Multimodal Structural Distribution of the p53 C Terminal Domain upon Binding to S100B via a Generalized Ensemble Method From Disorder to Extradisorder Journal of Chemical Theory and Computation 15 4 2597 2607 doi 10 1021 acs jctc 8b01042 PMID 30855964 S2CID 75138292 Kurcinski M Kolinski A Kmiecik S June 2014 Mechanism of Folding and Binding of an Intrinsically Disordered Protein As Revealed by ab Initio Simulations Journal of Chemical Theory and Computation 10 6 2224 2231 doi 10 1021 ct500287c PMID 26580746 Ciemny MP Badaczewska Dawid AE Pikuzinska M Kolinski A Kmiecik S January 2019 Modeling of Disordered Protein Structures Using Monte Carlo Simulations and Knowledge Based Statistical Force Fields International Journal of Molecular Sciences 20 3 606 doi 10 3390 ijms20030606 PMC 6386871 PMID 30708941 Garaizar A Espinosa JR September 2021 Salt dependent phase behavior of intrinsically disordered proteins from a coarse grained model with explicit water and ions The Journal of Chemical Physics 155 12 125103 Bibcode 2021JChPh 155l5103G doi 10 1063 5 0062687 PMID 34598583 S2CID 238249229 Uversky VN 2013 Digested disorder Quarterly intrinsic disorder digest January February March 2013 Intrinsically Disordered Proteins 1 1 e25496 doi 10 4161 idp 25496 PMC 5424799 PMID 28516015 Costantini S Sharma A Raucci R Costantini M Autiero I Colonna G March 2013 Genealogy of an ancient protein family the Sirtuins a family of disordered members BMC Evolutionary Biology 13 60 doi 10 1186 1471 2148 13 60 PMC 3599600 PMID 23497088 External links editIntrinsically disordered protein at Proteopedia MobiDB a comprehensive database of intrinsic protein disorder annotations IDEAL Intrinsically Disordered proteins with Extensive Annotations and Literature Archived 2020 05 02 at the Wayback Machine D2P2 Database of Disordered Protein Predictions Gallery of images of intrinsically disordered proteins First IDP journal covering all topics of IDP research IDP Journal Database of experimentally validated IDPs IDP ensemble database Archived 2018 03 10 at the Wayback Machine Retrieved from https en wikipedia org w index php title Intrinsically disordered proteins amp oldid 1182908919, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.