Báo cáo khoa học: Flexible nets The roles of intrinsic disorder in protein interaction networks potx

20 401 0
Báo cáo khoa học: Flexible nets The roles of intrinsic disorder in protein interaction networks potx

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

MINIREVIEW Flexible nets The roles of intrinsic disorder in protein interaction networks A Keith Dunker1, Marc S Cortese1, Pedro Romero1,2, Lilia M Iakoucheva3 and Vladimir N Uversky1,4 Department of Biochemistry and Molecular Biology, and the Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA School of Informatics, Indiana University – Purdue University Indianapolis, IN, USA Laboratory of Statistical Genetics, The Rockefeller University, New York, NY, USA Institute for Biological Instrumentation, Russian Academy of Sciences, Moscow Region, Russia Correspondence A.K Dunker, Department of Biochemistry and Molecular Biology, and the Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 714 N Senate Ave, Suite 250, Indianapolis, IN 46202, USA E-mail: kedunker@iupui.edu (Received 25 May 2005, revised 23 August 2005, accepted 30 August 2005) doi:10.1111/j.1742-4658.2005.04948.x Proteins participate in complex sets of interactions that represent the mechanistic foundation for much of the physiology and function of the cell These protein–protein interactions are organized into exquisitely complex networks The architecture of protein–protein interaction networks was recently proposed to be scale-free, with most of the proteins having only one or two connections but with relatively fewer ‘hubs’ possessing tens, hundreds or more links The high level of hub connectivity must somehow be reflected in protein structure What structural quality of hub proteins enables them to interact with large numbers of diverse targets? One possibility would be to employ binding regions that have the ability to bind multiple, structurally diverse partners This trait can be imparted by the incorporation of intrinsic disorder in one or both partners To illustrate the value of such contributions, this review examines the roles of intrinsic disorder in protein network architecture We show that there are three general ways that intrinsic disorder can contribute: First, intrinsic disorder can serve as the structural basis for hub protein promiscuity; secondly, intrinsically disordered proteins can bind to structured hub proteins; and thirdly, intrinsic disorder can provide flexible linkers between functional domains with the linkers enabling mechanisms that facilitate binding diversity An important research direction will be to determine what fraction of protein– protein interaction in regulatory networks relies on intrinsic disorder Scale-free networks and hubs In biological systems, processes such as growth, energy generation, cell division and signaling are integrated by large, intricate networks These biological networks, as well as certain nonbiological networks, especially those involved in communications such as the internet and cellular phone systems, are classified as scale-free networks (SFNs) [1–3] The basic feature that separates these networks from non-SFNs such as regular Abbreviations CaM, calmodulin; Cdk, cyclin-dependent protein kinase; CKI, Cdk inhibitor protein; GSK3b, glycogen synthase kinase beta; ID, intrinsically disordered; MoRE, molecular recognition element; NER, nucleotide excision repair; PDB, Protein Data Bank; PONDRÒ, predictors of naturally disordered regions; RGN, regular network; RNN, random network; SFN, scale-free network; XPA, xeroderma pigmentosum group A protein; FRAT, frequently rearranged in advanced T-cell lymphomas; Wnt, wingless type MMTV integration site family; HMG, high mobility group; VL-XT, a predictor of intrinsic disorder that integrates various methods-based predictor of long disordered regions and X-ray based N- and Cterminal predictors; VSL1, length-dependent predictor of intrinsic protein disorder; RPA, replication protein A; ERCC1, exchange repair cross complementing complex 1; TFIIH, transcription factor IIH; XAB, XPA binding protein; p27Kip, cyclin-dependent kinase inhibitor protein p27/1B FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS 5129 Flexible nets A K Dunker et al networks (RGNs) or random networks (RNNs) is the presence of hubs Hubs are highly connected nodes that have hundreds, thousands or even millions of links [1,4] The existence of hubs and their frequency impart two features to SFNs that provide substantial benefit to large complex networks: (a) increased robustness with regard to random defects and (b) shorter distances (in terms of the number of intervening nodes) between any two points [5] RGNs are grid-like with invariant node connectivity, whereas RNNs are characterized by stochastic variations in node connectivity [5,6] Despite the random placement of links in RNNs, the vast majority of nodes still have approximately the same connectivity A C 5130 Figure 1A,B compare an RNN to a similar-sized SFN to illustrate an important property of the latter In SNFs, the hub nodes are connected to a dramatically greater fraction of all nodes than the nodes with high connectivity in RNNs [7] This provides the ability for a signal to travel from any node to any other by traversing a small number of intervening nodes This feature imparts the so called small world property to SFNs [4,8] Extending the example to a biological context, Fig 1C represents an experimentally derived Saccharomyces cerevisiae protein–protein interaction map with 1870 protein ‘nodes’ connected by 2240 direct physical interactions [9] Visual comparison of the model SFN (Fig 1B) with the experimentally B Fig Comparison of simulated and actual protein interaction networks (A) The random network is rather homogeneous as most nodes have approximately the same number of links (Reproduced from [205] with the permission of the author, ª Institute of Physics and IOP Publishing Ltd., 2000–05.) (B) A scale-free network (SFN) is extremely inhomogeneous; while the majority of nodes have one or two links, a few nodes have a large number of links To show this, five nodes with the highest number of links are colored red, and their first neighbors are colored green While in the random network only 27% of the nodes are reached by the five most connected nodes, in the SFN more than 60% are This demonstrates the key role that hubs play in the SFN Note that both networks contain the same number of nodes (130) and links (430) (Reproduced from [205] with the permission of the author, ª Institute of Physics and IOP Publishing Ltd., 2000–05.) (C) Yeast protein interaction network map The color of a node signifies the phenotypic effect of removing the corresponding protein (red, lethal; green, nonlethal; orange, slow growth; yellow, unknown) (Modified from [9] with the permission of the authors, ª Nature Publishing Group, 1998–2005) FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al derived protein–protein interaction network (Fig 1C) suggests a similar underlying architecture The scale-free nature of protein–protein interaction networks gives them the advantages of high connectivity and robustness For example, the connectedness of RNNs decays steadily as nodes fail in a random fashion The surviving network breaks into progressively smaller and increasingly separate subnets that lose the ability to communicate with one another On the other hand, SFNs show much less degradation from random node failure because highly connected hubs serve to maintain the integrity of the network Because SFN hubs comprise a small fraction of total nodes, they are statistically less likely to fail as a result of random deleterious events Although the error tolerance of SFNs is high, it is important to note that the failure of hubs quickly leads to the breakdown of connectivity [7] This suggests that the SFNs are particularly resistant to random node removal but are extremely sensitive to targeted hub removal In agreement with these observations, analysis of the S cerevisiae protein–protein interaction network revealed that although proteins with five or fewer links constitute about 93% of the total number of proteins, only about 21% of them are essential to cell survival By contrast, only 0.7% of yeast proteins are known to have more than 15 links (i.e., hubs), but for 62% of these, deletion is lethal [9] A few caveats about the scale-free nature of biological networks are in order First, the network coverage of S cerevisiae [10,11], Caenorhabditis elegans [12], and Drosophila melanogaster [13] interactomes elucidated to date is sufficient only to suggest scale-free architecture That is, the examined networks appear to be scale-free in nature, but the data constitute only a small fraction of the full interactomes That the scale-free nature of biological protein–protein interaction networks is currently only a prediction was demonstrated by Han et al [14] These collaborators constructed simulated networks with random, exponential, scale-free and truncated normal topologies and a range of average connectivities When the four topologies were sampled at the level comparable to that used to construct current protein–protein interaction maps, there was insufficient distinction among the derived sample networks to unequivocally assign the underlying architectures Thus, conclusive proof that biological protein–protein interaction networks maintain a scale-free nature throughout full interactomes remains to be verified Secondly, experimental protein–protein interaction data contains a significant fraction of false negatives and positives This has been illustrated by studies comparing the results from various large-scale efforts [15– 18] Increasing the quality of existing data can be FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS Flexible nets addressed by comparing and combining datasets and adding additional methods of analysis such as gene neighborhood, gene co-occurrence, gene fusion events, mRNA expression correlations and lethality of knockouts [19] Despite the above described limitations, analysis of existing protein–protein interaction data can lead to useful information Comparison of protein–protein interaction networks across species has significant potential for the study of molecular evolution Useful tools have been constructed for the comparison of protein–protein interaction networks across species [20–23], and interesting evolutionary inferences are being made, such as the observation that proteins having a more central position in the networks of different species (i.e., hubs) appear to evolve more slowly and are evidently more likely to be essential for survival These observations are consistent with Fisher’s classic proposal that pleiotropy constrains evolution [24,25] Other important considerations include the timing and the locations of the hub protein interactions Some hub proteins have multiple simultaneous interactions (party hubs), while others have multiple sequential interactions separated in time or in space (date hubs) [26] It has been suggested that date hubs organize the proteome, connecting biological processes (modules; [27]) to each other, whereas party hubs act inside functional modules [26] and thereby may form scaffolds for various molecular machines or coordinated processes Neither the classical lock-and-key [28] nor the original induced fit mechanism [29] readily accommodates the multiple interactions of hub proteins, especially at higher connectivities Therefore, the existence of highly connected hubs in scale-free protein–protein interaction networks suggests a different mechanism for molecular recognition In fact, highly connected proteins were suggested to have some special, perhaps even common, structural features that would endow them with the ability to carry out highly specific interactions with many different proteins [30] Gunasekaran et al postulated that the relatively large solvent-accessible surface areas of extended disordered proteins could provide the potential for large intermolecular surfaces with less impact on cell size than if the same surfaces were provided by structured proteins [31] Rather than maintaining a smaller cell size, the key advantage of intrinsic disorder may lie in providing the molecular basis for the existence, flexibility, and evolution of interaction networks The following section explores a novel protein-disorder based mechanism that could provide the structural features necessary to allow proteins to carry out highly specific interactions with multiple, structurally diverse partners 5131 Flexible nets Many proteins have been shown to exist under apparently physiological conditions as dynamic ensembles Instead of having relatively fixed bonds and angles as in structured proteins, the backbone bonds and angles of such proteins vary significantly over time, with no specific equilibrium values while undergoing noncooperative conformational changes In other words, such proteins or protein regions not have rigid 3D structure under physiological conditions in vitro [31–47] Furthermore, these intrinsically disordered (ID) proteins and regions are known to carry out numerous biological functions including cell signaling [35], molecular recognition [48], and various other interactions with proteins and nucleic acids [32,34,35,37,39–43,49–51] Recently, a number of groups have published predictors of protein disorder, several of which are available on the web (reviewed in [48]; see also http://www disprot.org) These predictors are based on the assumption that the absence of rigid structure is encoded in specific features of the amino acid sequence [52,53] In fact, statistical analysis shows that amino acid sequences encoding for ID proteins or regions are significantly different from those of ordered proteins on the basis of local amino acid composition, flexibility, hydropathy, charge, coordination number and several other factors [34,52,54–56] A signature of a probable ID region is the presence of low sequence complexity coupled with amino acid compositional bias, characterized by a low content of bulky hydrophobic amino acids (Val, Leu, Ile, Met, Phe, Trp and Tyr), which typically form the cores of folded globular proteins, and a high proportion of particular polar and charged amino acids (Gln, Ser, Pro, Glu, Lys and, on occasion, Gly and Ala) [57,58] These attributes were used to train artificial neural networks using standard back propagation methods to develop a series of ‘predictors of naturally disordered regions’ (PONDRÒs) [52,55,56,59,60] That intrinsic protein disorder is a common phenomenon is illustrated by the fact that thousands of proteins in the Swiss-Prot database were identified by PONDRÒ as having long regions of sequence that share distinguishing sequence attributes with known ID regions [55] Furthermore, computational studies revealed that eukaryotes exhibit more disorder than either prokaryotes or archaea [34,60–62] For example, in 22 bacterial and seven archaebacterial proteomes, the percentage of proteins with predicted regions of disorder ranged from 7% to 33% and from 9% to 37%, respectively In contrast, in five eukaryotes, disorder ranged from 36% to 63% [60] This large jump in the percentage of proteins with long predictions of disorder in nucleated, rather 5132 A K Dunker et al than non-nucleated, organisms was both remarkable and unexpected To explain this and similar observations, it was hypothesized that the higher abundance of intrinsic disorder in eukaryotes could be a consequence of the increased need for cell signaling and regulation in higher organisms [34,35,58,60,63,64] Qualitatively, it seems reasonable that unstructured proteins could serve as hubs, providing a simpler basis for responding to changes in the environment as compared to rigid proteins For example, disordered regions can bind partners with both high specificity and low affinity [65], suggesting that disorder-based signaling and regulatory interactions can be highly specific but be easily reversed These capabilities meet the fundamental requirements of signaling interactions – specificity and reversibility [49] with minimal structural requirements Another crucial property of ID proteins and regions that could contribute to their function in signaling networks is binding diversity; i.e., their ability to partner with many other proteins and other ligands, such as nucleic acids [66] This opens the possibility of disordered regulatory regions that are capable of binding many different partners An interesting consequence of the capability of ID proteins and regions to interact with different binding partners is the potential for polymorphism in the bound state That is, such proteins could have completely different geometries in the rigidified structures that are induced upon binding to different partners [48] This conjecture has been confirmed at atomic resolution Portions of axin and frequently rearranged in advanced T-cell lymphoma protein (FRAT), which possess negligible sequence similarity, both interact with an intrinsically disordered loop of glycogen synthase kinase beta (GSK3b) that adopts ordered structure upon binding [67] The binding sites for the two molecules on GSK3b overlap substantially in the crystal structures solved for the axin–GSK3b and FRAT–GSK3b complexes [67] Furthermore, although both bound peptides are primarily helical, their detailed structures and interactions with GSK3b have substantial differences [67] The ability of GSK3b to bind two different proteins with high specificity via the same binding site is mediated by the conformational plasticity of the loop formed by residues 285–299 In the nonbound form of GSK3b [67], this loop is poorly defined in the electron density map, suggesting that it very likely occupies multiple conformations However, this loop is induced to accommodate one of two completely distinct well-ordered structures, each of which is specific to the bound partner [67,68] While some residues in this versatile GSK3b binding site are involved in interactions with both axin and FRAT, FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al their local conformations in the bound state are different In addition, other residues are involved uniquely with one ligand or the other [67] By extending such detailed observations to protein– protein interaction networks in general, we suggest that a unique feature of disordered regions in hub proteins could be structural plasticity in the unbound state, which when combined with the capability of interacting with multiple structurally distinct partners, results in structural polymorphism in the bound state These features have very important functional implications By this means, ID hub proteins and regions could serve multiple and distinct signaling networks and be regulated via different pathways For example, GSK3b plays a crucial role in the wingless-type MMTV integration site family (Wnt) signaling pathway by controlling the levels of b-catenin [69–71], and GSK3b is also known to be involved in insulin and growth factor signaling pathways [72–75] GSK3b functions as a signal transducer for these two completely independent pathways without any obvious cross-talk or interference [67] In the Wnt signaling network, a subset of the cellular GSK3b pool is incorporated into a multiprotein complex that brings GSK3b and its b-catenin substrate into close proximity In the insulin signaling pathway, GSK3b operates via a completely different mechanism, where the phosphorylation of Ser9 converts the disordered N-terminus of GSK3b to an autoinhibitory segment that blocks access to the active site substrate binding cleft [76] The functional segregation of the insulin and Wnt signaling networks requires either the absence of an exchange between the subsets of the cellular GSK3b molecules involved in each pathway, or suggests mutual exclusion of the two processes That is, the involvement of GSK3b with the axin–adenomatous polyposis coli complex can reverse (via the action of the specific phosphatases associated with the mentioned complex [77,78]) or override the inhibitory Ser9 phosphorylation present on a recruited GSK3b molecule via the substantial enhancement in activity towards b-catenin afforded by the axin ‘scaffolding’ [76] Importantly for this minireview, GSK3b uses two different ID regions to participate in two completely unrelated pathways: the disordered N-terminal fragment (residues 1–34) for insulin signaling and the disordered loop (residues 285–299) for the Wnt network Intrinsic disorder and protein–protein interaction networks The advantages of ID proteins and regions for forming associations with multiple partners led us to search the FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS Flexible nets literature for hub proteins having detailed structural characterization Table contains a few examples of structurally characterized hub proteins that were identified The range of structural types fell into three broad classes (as indicated in the ‘Percentage disordered’ column): entirely or mostly disordered (that is, nearly 100% disordered), partially disordered (a midrange percentage of disorder) and entirely or mostly ordered (nearly 0% disordered) For the mostly ordered hub proteins, we paid specific attention to the structure of the binding partners In many cases, these partners contained regions of intrinsic disorder One example of an ordered hub, calmodulin, makes use of a flexible hinge to facilitate binding diversity Each of these classes and their roles in protein–protein interaction networks will be discussed in turn Mostly disordered hubs Several hub proteins have been shown to be completely or almost completely disordered in solution, including a-synuclein, caldesmon, high mobility group protein A (HMGA), and synaptobrevin (Table 1) An interesting, well-studied, illustrative example of this group of hubs is provided by HMGA [formerly called HMG-I(Y)], a founding member of a new protein class called architectural transcription factors [79] As discussed in more detail below, this protein has been implicated in the development of cancer and several other pathological conditions [80] HMGA is considered a central hub of nuclear function, being able to bind to at least 18 known protein partners as well as to several specific DNA structures [80] Both circular dichroism (CD) [81] and nuclear magnetic resonance (NMR) [82,83] indicate that HMGA lacks structure, with the molecule exhibiting a random coil-like structure over its entire length The atypical electrophoretic mobility of this molecule [84] also suggests a high content of extended structure Figure 2A compares the results of PONDRÒ analysis by two predictors of intrinsic disorder, firstly, a predictor of intrinsic disorder that integrates various methods-based predictor of long disordered regions and X-ray based Nand C-terminal predictors (VL-XT) [52,57,59] and secondly, a length-dependent predictor of intrinsic protein disorder (VSL1) [85] While VL-XT is the most wellcharacterized member of the PONDRÒ family, VSL1 is more accurate overall and, indeed, obtained the best results of the 20 order ⁄ disorder predictors tested in the 6th Critical Assessment of Methods for Protein Structure Prediction (CASP6; http://predictioncenter.org/) In complete agreement with the experimental data, the predictor outputs in Fig 2A indicate that the HMGA 5133 5134 1–140 [154] 1–771 [157] 1–107 [161] 1–96 [163]) 170–1649 [169] 1–102, 210–273 [113] 1–184 [172] 1–73 [176]; 183–188, 224–227 [177]; 291–312 [178]; 319–323, 357–360 [179] 1–17 [181]; 17–24, 110– 125 [182]; 210–304 [183] 1–13, 390–414, 469–521 [185] a-synuclein (P37840) Caldesmon (P12957) HMGA (P17096) Synaptobrevin (P63027) BRCA1 (P38398) XPA (P23025) Estrogen receptor a (P03372) p53 (P04637) 77–81 [136] Actin (P68133) Calmodulin (P62152) a 1–7, 42–52 [195]a Cdk2 (P24941) 7⁄0 16 ⁄ 4⁄0 12 ⁄ 34 ⁄ 11 ⁄ 1⁄0 199 ⁄ 17 158 ⁄ 14 27 ⁄ 69 ⁄ 1900 ⁄ 40 95 ⁄ 451 ⁄ 24 13 ⁄ 11 ⁄ 3⁄0 7359 ⁄ 457 322 ⁄ 1⁄0 1⁄0 485 ⁄ 33 29 ⁄ N.D N.D DIP (inter ⁄ comp.) 10 ⁄ 235 ⁄ 35 BIND (inter ⁄ comp.) 2962 ⁄ 88 Disordered regions were based on missing residues in the PDB entries listed in the Swiss-Prot entry 36–46, 152–162 [121]a 14-3-3¢n (P63104) 12 16 26 29 31 63 79 100 100 100 100 Percent disordered (%) 68–77, 134–137, 230–245 [191]a Calcineurin, subunit A (Q08209) Mdm2 (Q00987) Disordered regions (start-stop) Protein (accession number) 33 125 97 31 72 239 116 41 119 27 27 STRING (inter.) 50 ⁄ 19 ⁄ 30 ⁄ 15 61 ⁄ 5⁄1 29 ⁄ 164 ⁄ 90 ⁄ 12 ⁄ 76 ⁄ 6⁄0 13 ⁄ 27 ⁄ 7⁄0 HPRD (inter ⁄ comp.) NFAT [186]; calcipressin [187]; cabin [188]; SOCS-3 [189]; calsarcin [190] p53, Wee1, Tau, Raf-1 Cdc25C, Bad [192] protein phosphatase A, cyclin E1, DNA polymerase alpha [193]; BRCA1, cyclin A [194] profilin [196]; deoxyribonuclease I, vitamin D binding protein, thymosin beta-4, cofilin [197] neurogranin, calcineurin, AC1 [198]; calponin [199]; caldesmon [200] p53, ARF, ATM, CK2, HIF-1a [184] parkin, tau, Ab [155]; 14-3-3, CaM [156] ERK [158]; S100a & b [159]; myosin, actin, calmodulin [160] AP1, NF-jB, C ⁄ EBPb, Oct )1 [80]; HIPK2, Sp1 [162] syntaxin A & 1B [164]; BAP31 [165]; VAMP assoc protein A [166]; VAMP assoc protein B [167]; SNAP-25 [168] p53, ATM, BRCA2, c-Myc [169]; Chk1 & [170] RPA70, RPA34, ERCC1, TFIIH, XAB1 & [171] p53 [173]; BRCA1 [174]; TATA box binding protein [172]; calmodulin, c-Jun [175] Mdm2, ATM, ERK, p38, BCL-XL [180] Illustrative partners Table Number of interacting protein partners for hub proteins Experimentally characterized disorder is described in terms of the start and stop residues of the disordered region(s) and overall percentage of disordered residues in the whole protein The BIND, DIP and HPRD database query results are presented as number of protein–protein interactions ⁄ number of complexes (inter ⁄ comp.), while the STRING search results are presented as number of protein–protein interactions (inter.) only Proteins are ordered by percentage of disorder BIND, the Biomolecular Interaction Network Database (http://bind.ca/) [201] DIP, the Database of Interacting Proteins (http://dip.doe-mbi.ucla.edu/) [202] STRING, Search Tool for the Retrieval of Interacting Genes ⁄ Proteins (associations with confidence scores > 0.7) (http://string.embl.de/) [203] HPRD, the Human Protein Reference Database (http://www.hprd.org/) [204] N.D., not in database Flexible nets A K Dunker et al FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al Flexible nets A Fig Order ⁄ disorder predictions on three hub proteins (A) PONDRÒ VL-XT (red) and VSL1 (magenta) predictions on the human HMGA protein sequence (Swiss-Prot accession number P17096) Green horizontal bars correspond to the areas of the protein that have been identified as the minimal regions required for specific protein–protein interactions with other transcription factors (after [80]): 1, IRF-1; 2, ATF-1 ⁄ c-Jun; 3, NF-Y; 4, SRF; 5, NF-jB; 6, p50; 7, Tst-1 ⁄ Oct-6; 8, HIPK-2 Although only eight target proteins are shown here, it has been established that HMGA physically interacts with at least 18 transcription factors [80] Dark yellow horizontal bars correspond to the areas of the protein (known as AT-hooks) that are involved in DNA binding A PONDRÒ score ¼ 0.5 corresponds to a prediction of disorder (B) PONDRÒ VL-XT (red) and VSL1 (magenta) predictions on the Xenopus laevis XPA protein sequence (Swiss-Prot accession number P27088) Vertical red and blue bars correspond to the accessible and inaccessible trypsin digestion sites, respectively Notice how, for the most part, cut sites within predicted ordered regions are not cleaved by the trypsin Green horizontal bars correspond to the functional regions of XPA and reflect interactions sites with the following binding partners: 1, 34 kDa subunit of RPA [103,104]; 2, ERCC1 [106,107]; 3, 70 kDa subunit of RPA [103,104]; 4, TFIIH [108,109]; 5, XAB1 [101] Dark yellow horizontal bar corresponds to the minimal fragment of XPA known to interact with damaged DNA [110] (C) PONDRÒ VL-XT (red) and VSL1 (dark pink) predictions on the human Cdk2 sequence (Swiss-Prot accession number P24941) Horizontal green bars correspond to the regions of Cdk2 involved in the interaction with p27Kip1 (residues with ˚ atoms within A of atoms of p27Kip1 [122]) B C sequence has a high propensity for disorder over its entire length However, HMGA was shown to undergo disorder-to-order transitions upon binding to DNA or protein partners [83,86,87] For example, the DNAbinding regions of the HMGA assume a planar, crescent-shaped configuration called the ‘AT-hook’ when specifically bound to the minor groove of short AT-rich stretches of DNA [83,86,87] Despite its lack of ordered structure, HMGA participates in a wide variety of nuclear processes ranging from modulation of chromosome and chromatin mechanics [88,89] to acting as an architectural tranFEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS scription factor that regulates the expression of more than 45 different eukaryotic and viral genes in vivo [79,90] Besides their association with whole chromosomes [88,89], HMGA proteins also bind to individual nucleosomes, both in vitro and in vivo [91,92] In addition to these unique DNA-binding characteristics, at least 18 different transcription factors have been reported to specifically interact with HMGA proteins (summarized in [80]) The list of proteins known to interact with HMGA includes transcription factors such as AP-1, ATF-2 ⁄ c-Jun heterodimer, IRF-1, c-Jun, NF-jB p50 ⁄ p65 heterodimer, C ⁄ EBPb, Elf-1, NF-AT, NF-jB 5135 Flexible nets p50 homodimer, NF-jB p65, NF-Y, Oct-1, Oct-2 A, PIAS3, Pu.1, RNF4, SRF, and Tst-1 ⁄ Oct-6 heterodimer [80] HMGA gene expression is maximal during embryonic development [93] and has been suggested to be involved in the control of cell growth and differentiation [94] Interestingly, overproduction of HMGA can be oncogenic and promote tumor progression and metastasis via dramatic alterations in numerous signaling pathways [80] Based on these observations, it was suggested that HMGA proteins function in the cell as highly connected ‘nodes’ of protein–DNA and protein–protein interactions that influence a diverse array of normal biological processes including growth, proliferation, differentiation and death [80] In summary, HMGA is a well-studied hub protein that in the absence of binding partners has been characterized to be completely disordered by NMR [82,83] and CD [81], supporting the hypothesis that hub proteins are strong candidates to possess appreciable amounts of disorder The HMGA example was also a major factor leading to the suggestion that hub proteins as a group might depend on intrinsic disorder [49] This supposition is supported below by several additional examples of hub proteins that utilize ID regions in their associations with multiple partners Partially disordered hub proteins Table also lists several hub proteins that possess an intermediate range of both ordered and disordered segments (internal loops and ⁄ or terminal tails), including BRCA1, Mdm2, XPA, p53, estrogen receptor a, and subunit A of calcineurin Disordered regions appear to play important roles in the binding interactions of these hub proteins The xeroderma pigmentosum group A protein (XPA) represents an interesting example of a partially disordered hub protein Xeroderma pigmentosum is an autosomal recessive human disease characterized by hypersensitivity to sunlight and a high incidence of skin cancer on sunexposed skin [95,96] This hypersensitivity is caused by defects in the nucleotide excision repair (NER) pathway that is necessary to correct many types of DNA damage [95,97] XPA, consisting of 273 amino acid residues, participates in the assembly of the damage recognition ⁄ incision complex, recruiting several other functional proteins to the site of damage [98] Particularly, it has been shown that this protein binds to three NER factors; replication protein A (RPA), exchange repair cross complementing complex (ERCC1) and transcription factor IIH (TFIIH), as 5136 A K Dunker et al well as to UV- or chemical carcinogen-damaged DNA [99,100] Furthermore, XPA was shown to interact with the cytoplasmic GTPase XPA-binding protein (XAB1) [101] and with a tetratricopeptide repeat protein XAB2 [102] Functionally, XPA can be divided into several distinct regions (Fig 2B): (a) the N-terminal fragment (residues 4–29) that binds to a 34 kDa subunit of RPA [103,104]; the basic amino acid region (residues 30–42) that is responsible for localizing XPA in the nucleus [105]; the acidic region (residues 78–84) that is important for interaction with ERCC1 [106,107]; and the C-terminal region (residues 226–273) that binds to TFIIH [108,109] Furthermore, the central region (residues 98–219) is the minimal polypeptide that preferentially binds damaged DNA [110] Finally, the fragment 98–187 is necessary for binding to the 70 kDa subunit of RPA [103,104] Figure 2B presents the distribution of the PONDRÒ VL-XT and VSL1 scores within the XPA sequence and illustrates the long predicted disordered regions at or near the two ends (from M1 to A55 and from S63 to P88 at the N-terminus and from L183 to E230 near the C-terminus) Importantly, Fig 2B shows that the central DNA-binding domain is likely to be mostly ordered, whereas the multiple protein binding sites are located in the regions that are likely to be disordered In agreement with the predictions (Fig 2B), NMR solution structure of a human XPA fragment containing the minimal DNA-binding domain (residues 98–219), revealed that one-third of this molecule is disordered [111,112] These conclusions were further confirmed by the results of limited proteolysis analysis [113]: mild trypsin digestion produced cuts at 33 of the possible 48 sites, with no cleavage at any of the 14 possible sites in the internal DNA-binding region (Q85–I179) (Fig 2B) The observed cleavage sites were predominantly in two of the large regions of predicted disorder [113] In general, it is believed that cut sites within disordered regions are more easily cleaved by proteases than those found in structured areas [114,115] Thus, excellent agreement was observed between PONDRÒ prediction of order and disorder and the observed sites of protease resistance and sensitivity, respectively [113] Summarizing, XPA is an illustrative example of the class of hub proteins that contain disordered tails and ⁄ or loops as well as ordered regions Importantly, flexibility of the disordered fragments in such proteins facilitates interactions with multiple binding partners without sacrificing specificity and enhances the ability of hub proteins to participate in multiple signaling pathways FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al Ordered hubs interacting with disordered partners Table lists four examples of protein hubs that are mostly ordered: actin, calmodulin, Cdk2, and 14-3-3¢n Below, we describe how ordered hubs may interact with intrinsically disordered binding partners, and how such interactions may play crucial roles in regulation and coordination of hub protein activities The orderly progression of cells through the various phases of the cell cycle is governed by several distinct cyclin-dependent protein kinases (Cdks), which therefore are considered as the master timekeepers of cell division [116] Unlike other protein kinases, Cdks are regulated by binding to their cyclin protein partners, forming active heterodimeric complexes Eight Cdk family members (Cdk1–Cdk8) and nine cyclins (A–I) have been identified so far Interestingly, each Cdk pairs with a separate cyclin class, most of which have at least two members [117,118] For example, Cdk1 together with cyclin B1 directs the G2 ⁄ M transition Exit from G1, in contrast, is primarily under the control of cyclin D ⁄ Cdk4 ⁄ Finally, two other cyclins (A and E) that pair with Cdk2 are required for the G1 ⁄ S transition and progression through the S phase [117,118] All Cdks have similar sizes (30–40 kDa) and share at least 40% sequence identity, including the highly conserved 300 residue catalytic core On the contrary, the cyclin subunits vary in size (30–80 kDa), but all contain a homologous 100 residue cyclin box domain The Cdk subunits are not catalytically active unless they bind to a cyclin partner and form a basally active complex The fully active complex is produced when the Cdk is phosphorylated [116,119] Crystal structures of several Cdks (phosphorylated and dephosphorylated) and their complexes with cyclins and inhibitors have been solved [119] All Cdks have the same overall fold as other eukaryotic protein kinases For example, monomeric Cdk2 consists of an N-terminal lobe rich in b-sheet (N lobe), a larger C-terminal lobe rich in a-helix (C lobe), and a deep cleft at the junction of the two lobes that is the site of ATP binding and catalysis [120] Figure 2C represents distribution of the PONDRÒ VL-XT and VSL1 scores within the human Cdk2 sequence and illustrates that this protein is likely to be almost completely ordered, having only several small regions of predicted disorder The crystal structure of human Cdk2 (PDB ID: 1URW) is consistent with this prediction, with only two regions containing missing residues [121] Interestingly, these two segments, residues 36–46 and 152–162, overlap with experimentally identified regions of Cdk2 that interact with cyclin-dependent kinase inhibitor protein p27/1B (p27Kip1) (Fig 2C) [122] FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS Flexible nets Regulation of Cdk activity is essential for the proper timing and coordination of numerous nuclear processes, including DNA replication and chromosome separation, required for cell growth and division Not surprisingly, the activity of Cdks throughout the cell cycle is precisely directed by a combination of several mechanisms, including the control of cycle-dependent variations in the levels of activating partners, cyclins (via regulation of their synthesis and degradation); coordination of Cdk phosphorylation and dephosphorylation, which is required for controlled activation and deactivation of Cdks; and variations in the levels of the Cdk inhibitor proteins, CKIs, responsible for the deactivation of the Cdk–cyclin complexes [116,119] Four major mammalian CKIs, which regulate cell proliferation through their physical interactions in the nucleus with Cdks, have been discovered so far p21Waf1 ⁄ Cip1 ⁄ Sdi1 and p27Kip1 inactivate Cdk2 and Cdk4 cyclin complexes by binding to them, whereas p16INK4 and p15INK4B are specific for Cdk4 and Cdk6 Importantly, CKIs are able to inhibit Cdk–cyclin activity by blocking formation of active Cdk–cyclin complexes via binding to inactive Cdk or by binding to the active complex [116,119] The Cdk inhibitor p21Waf1 ⁄ Cip1 ⁄ Sdi1 is important for p53-dependent cell cycle control, mediating G1 ⁄ S arrest through inhibition of Cdks and possibly through inhibition of DNA replication [123] A striking disorder-to-order transition for p21Waf1 ⁄ Cip1 ⁄ Sdi1 upon binding to one of its biological targets, Cdk2, was demonstrated by proteolytic mapping, CD spectropolarimetry, and NMR spectroscopy [66] In fact, it has been established that p21Waf1 ⁄ Cip1 ⁄ Sdi1 and its NH2-terminal fragment, being active as Cdk inhibitors, lacked stable secondary or tertiary structure in free solution However, the p21Waf1 ⁄ Cip1 ⁄ Sdi1 NH2-terminus adopts an ordered, stable conformation when bound to Cdk2 [66] This intrinsically disordered nature probably explains the ability of p21Waf1 ⁄ Cip1 ⁄ Sdi1 to bind to and to inhibit a diverse family of cyclin–Cdk complexes, including cyclin A–Cdk2, cyclin E–Cdk2, and cyclin D–Cdk4 [124] Thus, the intrinsic disorder of p21Waf1 ⁄ Cip1 ⁄ Sdi1 is associated with binding diversity and helps to explain the role for structural disorder in mediating binding specificity in biological systems [66] The p27Kip1 protein is another member of the p21 family of Cdk inhibitors that negatively regulate the cell cycle and thereby contributes to cellular growth and development [125,126] p27Kip1 contains an N-terminal Cdk-inhibition domain and a C-terminal domain of unknown function called the QT domain [125,127] The crystal structure of the human p27Kip1 Cdkinhibition domain (residues 22–106) bound to human cyclin A–Cdk2 complex shows that residues 25–93 of 5137 Flexible nets p27Kip1 bind in an ordered conformation comprising an a-helix, a 310 helix, and b-structure [120] Importantly, the p27Kip1 Cdk-inhibition domain was shown to lack an intramolecular hydrophobic core Instead, p27Kip1 interacts with the cyclin A–Cdk2 complex as an extended structure, being bound to both cyclin A and Cdk2 On cyclin A, it interacts with a groove formed by conserved cyclin box residues On Cdk2, p27Kip1 binds and rearranges the amino-terminal lobe and also inserts into the catalytic cleft, mimicking ATP in the context of the cyclin A–Cdk2 complex [120] In contrast, the unbound p27Kip1 Cdk-inhibition domain is intrinsically disordered (natively unfolded) as shown by both CD and NMR spectroscopy The NMR spectra lack chemical-shift dispersion and exhibit negative peaks for the 1H-15N heteronuclear nuclear Overhauser effect [122,128,129] Despite showing disorder before binding its targets, p27Kip1 has nascent, but transient, secondary structure that may have a function in molecular recognition [122] The kinetic analysis of p27Kip1 folding induced by its binding to Cdk2–cyclin A complex revealed that this is a sequential process initiated by binding to cyclin A, which is accompanied by folding of 34 residues (as estimated by a method suggested in [130]) The binding of p27Kip1 to Cdk2 leading to the inhibition of kinase activity is much slower and is accompanied by the folding of 59 residues [122] Based on these observations, it was proposed that p27Kip1 (and potentially other CKIs, such as p21 and p57) function essentially as ‘molecular staples’, wherein the ‘prongs’ of the staple (domains and 2) are unstructured and flexible before binding Cdk–cyclin complexes The staple analogy is completed by a linker helix (partially structured in the unbound state) that connects the two prongs This analogy is illustrated in Fig 3A, which presents a model for p27Kip1 binding to the Cdk2–cyclin A binary complex and shows the importance of both preformed, but transient structure in the linker region and the flexible nature of domains and for the efficient functioning of this protein [122] Figure 3B shows that p27Kip1 is predicted to be mostly disordered by both PONDRÒ VLXT and VSL1 Importantly, a region of predicted order overlaps with the fragment of p27Kip1 shown to contain a significant amount of regular secondary structure in its complex with Cdk2–cyclin A The above-mentioned sequential folding-upon-binding mechanism has been suggested to be crucial for the selective inhibition of specific Cdk–cyclin complexes by corresponding CKIs Furthermore, p21Waf1 ⁄ Cip1 ⁄ Sdi1 and p27Kip1 target the cell cycle CDKs (Cdk1, Cdk2, Cdk3, Cdk4 and Cdk6) but fail to bind and inhibit Cdk5 and Cdk7 [124] Although the surfaces of the Cdks 5138 A K Dunker et al are not markedly different from each other and therefore cannot provide a basis for specificity, important differences in surface residues of the cyclin partners of these Cdks have been described [122] Thus, p27Kip1 and other CKIs may have evolved to first recognize and bind these specificity-determining sites and then to fold into an extended structure that extends to the kinase subunit and consequently inhibits its activity [122] Flexible linkers between functional domains Calmodulin (CaM) is a 148 residue protein with four calcium binding sites that serves to mediate extracellularly induced Ca2+ signaling within the cytosol CaM modulates the activity of a large number of enzymes by direct binding, with both calcium-dependent [131] and calcium-independent [132] binding modes, with the former likely to be much more common than the latter The regions bound by CaM are typically about 20 residues in length and mostly a-helical in nature [133,134] CaM binding targets exhibit limited sequence identity and in many cases are nonhomologous, thus a mechanism that incorporates specificity but permits diversity must be encoded in CaM’s structure The X-ray crystal structure of CaM is dumbbelllike with two homologous globular domains connected by a rigid 26 residue a-helix [135] Subsequent to the X-ray structure determination, NMR analyses revealed that residues 77–81 in the middle of the central helix were highly flexible and functioned as a hinge [136] This hinge facilitates a binding mode in which CaM surrounds the target regions of its partners within the two Ca2+-binding, globular domains, and in some cases the hinge region remains unstructured after association with the CaM target [132] The interior faces of the globular domains have features that accommodate target diversity, such as nonrigid helix–helix packing that allow backbone adjustments and high methionine contents that are especially adept at side chain adjustments [132] An important structural feature enabling intermolecular binding with maximal surface area of interaction is the flexible connector between the two globular domains This flexibility accommodates a high diversity of sequence features in the target by allowing the CaM surface to seek complementary interactions by sampling different positions and orientations relative to the binding target surface Additionally, the flexible hinge facilitates variable separation of the two globular domains after binding has occurred, again allowing for binding diversity [136] Despite the small size of the disordered region (just five residues) and the slight FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al Flexible nets A Fig Disorder-to-order transition upon binding for p27Kip1 (A) A model for p27Kip1 binding to the Cdk2–cyclin A binary complex p27Kip1 is yellow, Cdk2 (K2) cyan and cyclin A (A) magenta In these panels, the subunit not present in the experimental binding reactions is gray to emphasize the relevance of experimental data for binary binding reactions to the mechanism of binding the Cdk2–cyclin A complex (right) (Modified from [122] with the permission of the authors, ª Nature Publishing Group, 1998–2005.) (B) PONDRỊ VL-XT (red) and VSL1 (dark pink) predictions on the human p27Kip1 sequence (Swiss-Prot accession number P46527) Green horizontal bars correspond to the regions of the protein involved in interaction with Cdk2 and cyclin A: 1, domain interacting with cyclin A; 2, a linker helix involved in binding both cyclin A and Cdk2; 3, domain interacting with Cdk2 [122] Blue and dark yellow horizontal bars correspond to the helices and b-structure stabilized by the formation of a triple complex, cyclin A–Cdk2–p27Kip1 B amount of disorder in the entire protein (just 3%), this disorder is crucial for the binding mechanism that enables CaM to associate with multiple partners Overall, a wide range of diverse sequences are recognized by CaM [133,134] ID regions vs dehydrons as the basis for binding to multiple partners The examples presented above emphasize the importance of intrinsic disorder for hub protein function: ID FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS regions provide hubs with the ability to bind multiple structurally diverse targets, thereby enabling them to participate in and possibly regulate multiple networks An alternative conjecture has been made, namely that the interactivity and connectivity of proteins of known structure in proteomic networks depends on the number of dehydrons (backbone hydrogen bonds that are insufficiently protected from the surrounding water molecules) observed for each protein [137–142] Dehydrons, the coulombic stabilizing energy of which increases as water is excluded, are effectively adhesive, 5139 Flexible nets because association-induced local water removal increases this hydrogen-bond stability [141] Important for the present work, the application of the dehydron concept in an analysis of the evolution of the yeast proteomic network [11,143], indicated that new binding partnerships could be promoted by relaxing the structure of the packing interface, which effectively increases the number of dehydrons [144] This was interpreted to mean that the rate of increase in interactome complexity (i.e., the rate that new connectivities are adopted) coincides with the rate of increase of the number of dehydrons According to this conjecture, proteins with greater numbers of dehydrons represent more plausible molecules for hub evolution How did the previous authors carry out the analysis of the yeast interactome and show that hub proteins were enriched in dehydrons? First, they showed that there is a statistical correlation between the local PONDRÒ VL-XT score and the degree of protection of the indicated hydrogen bond [144] Based on this correlation, PONDRÒ VL-XT scores were used to show that increases in the number of apparent dehydrons represented a plausible basis for SFN evolution [144] Although the occurrences of dehydrons and intrinsic disorder scores are correlated, it is not immediately evident which characteristic – dehydrons or disorder – actually explains the previous observations [144] and thus may provide the basis for hub evolution As reviewed in more detail here and elsewhere [40,48], disordered regions have been shown to be able to adopt different structures upon interaction with structurally diverse partners (thereby increasing the potential number of partners that can be accommodated by a given protein segment), but no evidence has yet been presented that dehydrons have a similar capability Thus, mutation-driven variation of locally disordered regions is more likely than dehydrons alone to be one of the key structural factors leading to the evolution of hub proteins How ID regions work? To explain one of the potential mechanisms used by ID proteins to interact with their binding partners, the concept of ‘molecular recognition element’ (MoRE) was introduced [145,146] The MoRE defines an interactionprone short segment of disorder that becomes ordered upon specific binding There are three basic types of MoREs: those that form a-helical structures upon binding; those that form b-strands (in which the peptide forms a b-sheet with additional b-strands provided by the protein partner); and those that form irregular structures when binding [48,145–147] Proposed names for these structures are a-MoRE, b-MoRE, and I-MoRE, 5140 A K Dunker et al respectively [48] Of course, a given MoRE could be composed of more than one type of secondary structure type when bound to its partner, resulting in complex MoREs, such as I-a-I-MoREs, a-b-MoREs, etc MoREs can be detected experimentally as segments of disordered regions that maintain some residual structure (as in the case of p27Kip1 [122]) They also can be discovered by analysis of protein–protein complexes deposited in the Protein Data Bank (PDB) [148] that consist of short nonglobular protein fragments bound to a large globular partner [48,145–147] Originally, 14 a-MoREs were described in 12 proteins [147] However, subsequent, more detailed analysis of PDB revealed more than 2500 such complexes, further analysis of which gave a nonredundant set of 372 nonhomologous MoREs This dataset includes  10 000 residues, 27% of which were assigned to a-helical conformation, 12% were b-structural and 48% were irregular, whereas the remaining 13% of the residues were disordered (based on missing coordinates in the PDB files) (A Mohan, P Romero, C.J Oldfield, V.N Uversky & A.K Dunker, unpublished data) Overall, this analysis shows that complexes of short nonglobular protein fragments bound to large globular proteins are rather abundant in PDB, and thus in nature Alternatively, MoREs can be detected computationally [146,147] In fact, for several proteins it was noted that PONDRÒ VL-XT could identify experimentally characterized binding regions as short downward spikes (signifying propensity for order) flanked by strong predictions of disorder [145,149,150] By combining additional sequence-derived criteria to the downward spike indications from PONDRÒ analysis, we developed an algorithm that identifies regions of high a-MoRE propensity [146,147] Application of this predictor to several genomes revealed that, while somewhat less than 3% of bacterial and archaea proteins contain regions of high a-MoRE propensities, about 23% of eukaryotic proteins in general, and almost 50% of eukaryotic regulatory proteins in particular, contain a-MoRE signals [146,147] This study suggests that disorder-to-helix transitions are common in protein interaction networks, but laboratory experiments are needed to test whether this mechanism is indeed as common as suggested Concluding remarks Systematic postgenome proteome-wide analysis of protein interactions using large-scale two-hybrid screens suggest that these interactions can be described as complex SFNs [9,26,151] On the other hand, many traditional approaches have been developed to analyze interactions, coordination, signaling and regulation on FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al a smaller scale (e.g within the scope of a single or multiple interacting pathways) Quite often these small-scale approaches yielded interesting results that were used to develop models that correctly predicted outcomes of changes and interruptions within the system studied Integrating small-scale network information with global network information represents an important but difficult task Successful completion of this task could lead to improved quality of the overall data, shed light on the mechanisms of timing and regulation, indicate how global and local properties of complex macromolecular networks affect observable biological properties (phenotype) and functions (physiology) and suggest how changes in such properties contribute to human diseases Several groups are productively working on the task of understanding interesting subnetworks [152,153] The example of GSK3b, in which different regions of intrinsic disorder are independently involved in different regulatory pathways, suggests that identifying and cataloguing ID regions in hub proteins could provide a useful approach for organizing protein–protein interaction subnetworks into larger networks We have presented evidence that ID proteins can play crucial functional roles in protein interaction networks This evidence indicates how some hubs carry out function, how some local nets are integrated into global networks, and how multiple processes can be coordinated, regulated, timed and isolated from one another The information available at this stage in the developing field of systems biology provides strong evidence for the existence of extensive complex networks combining global and local properties, but also demonstrates that much more data will be needed to develop reliable models Carrying out a comparison of the number of interactions involving at least one disordered partner with the number of interactions involving structured partners for a few selected subnetworks would be very useful Such a comparison would provide an estimate of the fraction of protein–protein interactions that utilize ID protein regions and the fraction that involve only structured proteins While we anticipate that disorder-to-order transitions might be very common in hub protein interactions because of the binding diversity advantage that disorder provides, the extracted ratio would provide an estimate of the frequency that this property is utilized on global scales Studies along these lines are currently in progress Acknowledgements We thank A.L Barabasi for permission to modify published figures and R.W Kriwacki for assistance FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS Flexible nets with figure modification We also thank Tanguy Le Gall and Molecular Kinetics, Inc for the use of the proprietary fragment-mapping program to mine PDB for some of the data presented in Table The Indiana Genomics Initiative (INGEN) and NIH Grant R01 LM007688-0A1 provided support to A.K.D This work was supported in part by INTAS 2001-2347 Grant to V.N.U., and L.M.I was supported by the grant MCB-0444818 from the National Science Foundation References Goh KI, Oh E, Jeong H, Kahng B & Kim D (2002) Classification of scale-free networks Proc Natl Acad Sci USA 99, 12583–12588 Jeong H, Tombor B, Albert R, Oltvai ZN & Barabasi AL (2000) The large-scale organization of metabolic networks Nature 407, 651–654 Barabasi AL & Albert R (1999) Emergence of scaling in random networks Science 286, 509–512 Watts DJ & Strogatz SH (1998) Collective dynamics of ‘small-world’ networks Nature 393, 440–442 Barabasi AL & Bonabeau E (2003) Scale-free networks Sci Am 288, 60–69 ´ Erdos P & Renyi A (1960) On the evolution of random ă graphs Publ Math Inst Hung Acad Sci 5, 17–61 Albert R, Jeong H & Barabasi AL (2000) Error and attack tolerance of complex networks Nature 406, 378–382 Milgram S (1967) The small world problem Psycol Today 2, 60–67 Jeong H, Mason SP, Barabasi AL & Oltvai ZN (2001) Lethality and centrality in protein networks Nature 411, 41–42 10 Ito T, Chiba T, Ozawa R, Yoshida M, Hattori M & Sakaki Y (2001) A comprehensive two-hybrid analysis to explore the yeast protein interactome Proc Natl Acad Sci USA 98, 4569–4574 11 Uetz P, Giot L, Cagney G, Mansfield TA, Judson RS, Knight JR, Lockshon D, Narayan V, Srinivasan M, Pochart P, Qureshi-Emili A, Li Y, Godwin B, Conover D, Kalbfleisch T, Vijayadamodar G, Yang M, Johnston M, Fields S & Rothberg JM (2000) A comprehensive analysis of protein–protein interactions in Saccharomyces cerevisiae Nature 403, 623–627 12 Li S, Armstrong CM, Bertin N, Ge H, Milstein S, Boxem M, Vidalain PO, Han JD, Chesneau A, Hao T et al (2004) A map of the interactome network of the metazoan C elegans Science 303, 540–543 13 Giot L, Bader JS, Brouwer C, Chaudhuri A, Kuang B, Li Y, Hao YL, Ooi CE, Godwin B, Vitols E et al (2003) A protein interaction map of Drosophila melanogaster Science 302, 1727–1736 5141 Flexible nets 14 Han JD, Dupuy D, Bertin N, Cusick ME & Vidal M (2005) Effect of sampling on topology predictions of protein–protein interaction networks Nat Biotechnol 23, 839–844 15 Bork P, Jensen LJ, von Mering C, Ramani AK, Lee I & Marcotte EM (2004) Protein interaction networks from yeast to human Curr Opin Struct Biol 14, 292– 299 16 Cesareni G, Ceol A, Gavrila C, Palazzi LM, Persico M & Schneider MV (2005) Comparative interactomics FEBS Lett 579, 1828–1833 17 von Mering C, Krause R, Snel B, Cornell M, Oliver SG, Fields S & Bork P (2002) Comparative assessment of large-scale data sets of protein–protein interactions Nature 417, 399–403 18 Bader GD & Hogue CW (2002) Analyzing yeast protein–protein interaction data obtained from different sources Nat Biotechnol 20, 991–997 19 Hoffmann R & Valencia A (2003) Protein interaction: same network, different hubs Trends Genet 19, 681– 683 20 Wu CH, Huang H, Nikolskaya A, Hu Z & Barker WC (2004) The iProClass integrated database for protein functional analysis Comput Biol Chem 28, 87–96 21 Huang TW, Tien AC, Huang WS, Lee YC, Peng CL, Tseng HH, Kao CY & Huang CY (2004) POINT: a database for the prediction of protein–protein interactions based on the orthologous interactome Bioinformatics 20, 3273–3276 22 Kelley BP, Yuan B, Lewitter F, Sharan R, Stockwell BR & Ideker T (2004) PathBLAST: a tool for alignment of protein interaction networks Nucleic Acids Res 32, W83–W88 23 von Mering C, Jensen LJ, Snel B, Hooper SD, Krupp M, Foglierini M, Jouffre N, Huynen MA & Bork P (2005) STRING: known and predicted protein–protein associations, integrated and transferred across organisms Nucleic Acids Res 33, D433–D437 24 Hahn MW & Kern AD (2005) Comparative genomics of centrality and essentiality in three eukaryotic protein interaction networks Mol Biol Evol 22, 803–806 25 Huang S (2004) Back to the biology in systems biology: what can we learn from biomolecular networks? Brief Funct Genomic Proteomic 2, 279–297 26 Han JD, Bertin N, Hao T, Goldberg DS, Berriz GF, Zhang LV, Dupuy D, Walhout AJ, Cusick ME, Roth FP & Vidal M (2004) Evidence for dynamically organized modularity in the yeast protein–protein interaction network Nature 430, 88–93 27 Hartwell LH, Hopfield JJ, Leibler S & Murray AW (1999) From molecular to modular cell biology Nature 402, C47–C52 28 Fischer E (1894) Einfluss der configuration auf die wirkung derenzyme Ber Dt Chem Ges 27, 2985–2993 5142 A K Dunker et al 29 Koshland DE Jr, Ray WJ Jr & Erwin MJ (1958) Protein structure and enzyme action Fed Proc 17, 1145–1150 30 Hasty J & Collins JJ (2001) Protein interactions Unspinning the web Nature 411, 30–31 31 Gunasekaran K, Tsai CJ, Kumar S, Zanuy D & Nussinov R (2003) Extended disordered proteins: targeting function with less scaffold Trends Biochem Sci 28, 81–85 32 Dunker AK, Brown CJ, Lawson JD, Iakoucheva LM & Obradovic Z (2002) Intrinsic disorder and protein function Biochemistry 41, 6573–6582 33 Dunker AK, Brown CJ & Obradovic Z (2002) Identification and functions of usefully disordered proteins Adv Protein Chem 62, 25–49 34 Dunker AK, Lawson JD, Brown CJ, Williams RM, Romero P, Oh JS, Oldfield CJ, Campen AM, Ratliff CM, Hipps KW et al (2001) Intrinsically disordered protein J Mol Graph Model 19, 26–59 35 Iakoucheva LM, Brown CJ, Lawson JD, Obradovic Z & Dunker AK (2002) Intrinsic disorder in cell-signaling and cancer-associated proteins J Mol Biol 323, 573–584 36 Uversky VN (2003) Protein folding revisited A polypeptide chain at the folding-misfolding-nonfolding cross-roads: which way to go? Cell Mol Life Sci 60, 1852–1871 37 Uversky VN (2002) Natively unfolded proteins: a point where biology waits for physics Protein Sci 11, 739– 756 38 Uversky VN (2002) What does it mean to be natively unfolded? Eur J Biochem 269, 2–12 39 Dyson HJ & Wright PE (2002) Coupling of folding and binding for unstructured proteins Curr Opin Struct Biol 12, 54–60 40 Dyson HJ & Wright PE (2005) Intrinsically unstructured proteins and their functions Nat Rev Mol Cell Biol 6, 197–208 41 Wright PE & Dyson HJ (1999) Intrinsically unstructured proteins: re-assessing the protein structure-function paradigm J Mol Biol 293, 321–331 42 Namba K (2001) Roles of partly unfolded conformations in macromolecular self-assembly Genes Cells 6, 1–12 43 Demchenko AP (2001) Recognition between flexible protein molecules: induced and assisted folding J Mol Recognit 14, 42–61 44 Tompa P (2002) Intrinsically unstructured proteins Trends Biochem Sci 27, 527–533 45 Tompa P (2003) Intrinsically unstructured proteins evolve by repeat expansion Bioessays 25, 847–855 46 Bracken C, Iakoucheva LM, Romero PR & Dunker AK (2004) Combining prediction, computation and experiment for the characterization of protein disorder Curr Opin Struct Biol 14, 570–576 FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al 47 Fink AL (2005) Natively unfolded proteins Curr Opin Struct Biol 15, 35–41 48 Uversky VN, Oldfield CJ & Dunker AK (2005) Showing your ID: intrinsic disorder as an ID for recognition, regulation and cell signaling J Mol Recognit 18, 343–384 49 Dunker AK & Obradovic Z (2001) The protein trinity – linking function and disorder Nat Biotechnol 19, 805–806 50 Uversky VN, Gillespie JR & Fink AL (2000) Why are ‘natively unfolded’ proteins unstructured under physiologic conditions? Proteins 41, 415–427 51 Daughdrill GW, Pielak GJ, Uversky VN, Cortese MS & Dunker AK (2005) Natively disordered proteins In Handbook of Protein Folding (Buchner J & Kiefhaber T, eds), pp 271–353 Wiley-VCH-Verlag GmbH KGaA, Weinheim, Germany 52 Romero P, Obradovic Z, Kissinger C, Villafranca JE & Dunker AK (1997) Identifying disordered regions in proteins from amino acid sequence Proceedings of International Conference on Neural Networks 1, 90–95 53 Obradovic Z, Peng K, Vucetic S, Radivojac P, Brown CJ & Dunker AK (2003) Predicting intrinsic disorder from amino acid sequence Proteins 53 (Suppl 6), 566– 572 54 Romero P, Obradovic Z & Dunker AK (1997) Sequence data analysis for long disordered regions prediction in the calcineurin family Genome Inform Ser Workshop Genome Inform 8, 110–124 55 Romero P, Obradovic Z, Kissinger CR, Villafranca JE, Garner E, Guilliot S & Dunker AK (1998) Thousands of proteins likely to have long disordered regions Pac Symp Biocomput 437–448 56 Dunker AK, Garner E, Guilliot S, Romero P, Albrecht K, Hart J, Obradovic Z, Kissinger C & Villafranca JE (1998) Protein disorder and the evolution of molecular recognition: theory, predictions and observations Pac Symp Biocomput 473–484 57 Romero P, Obradovic Z, Li X, Garner EC, Brown CJ & Dunker AK (2001) Sequence complexity of disordered protein Proteins 42, 38–48 58 Vucetic S, Brown CJ, Dunker AK & Obradovic Z (2003) Flavors of protein disorder Proteins 52, 573– 584 59 Li X, Romero P, Rani M, Dunker AK & Obradovic Z (1999) Predicting Protein Disorder for N-, C-, and Internal Regions Genome Inform Series Workshop Genome Inform 10, 30–40 60 Dunker AK, Obradovic Z, Romero P, Garner EC & Brown CJ (2000) Intrinsic protein disorder in complete genomes Genome Inform Series Workshop Genome Inform 11, 161–171 61 Oldfield CJ, Cheng Y, Cortese MS, Brown CJ, Uversky VN & Dunker AK (2005) Comparing and combining predictors of mostly disordered proteins Biochemistry 44, 1989–2000 FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS Flexible nets 62 Ward JJ, Sodhi JS, McGuffin LJ, Buxton BF & Jones DT (2004) Prediction and functional analysis of native disorder in proteins from the three kingdoms of life J Mol Biol 337, 635–645 63 Liu J & Rost B (2001) Comparing function and structure between entire proteomes Protein Sci 10, 1970– 1979 64 Sigler PB (1988) Transcriptional activation Acid blobs and negative noodles Nature 333, 210–212 65 Schulz GE (1979) Nucleotide binding proteins In Molecular Mechanism of Biological Recognition (Balaban M, ed.), pp 79–94 Elsevier ⁄ North-Holland Biomedical Press, New York 66 Kriwacki RW, Hengst L, Tennant L, Reed SI & Wright PE (1996) Structural studies of p21Waf1 ⁄ Cip1 ⁄ Sdi1 in the free and Cdk2-bound state: conformational disorder mediates binding diversity Proc Natl Acad Sci USA 93, 11504–11509 67 Dajani R, Fraser E, Roe SM, Yeo M, Good VM, Thompson V, Dale TC & Pearl LH (2003) Structural basis for recruitment of glycogen synthase kinase 3beta to the axin-APC scaffold complex Embo J 22, 494–501 68 Bax B, Carter PS, Lewis C, Guy AR, Bridges A, Tanner R, Pettman G, Mannix C, Culbert AA, Brown MJ, Smith DG & Reith AD (2001) The structure of phosphorylated GSK-3beta complexed with a peptide, FRATtide, that inhibits beta-catenin phosphorylation Structure (Camb) 9, 1143–1152 69 Ding Y & Dale T (2002) Wnt signal transduction: kinase cogs in a nano-machine? Trends Biochem Sci 27, 327–329 70 Polakis P (2000) Wnt signaling and cancer Genes Dev 14, 1837–1851 71 He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B & Kinzler KW (1998) Identification of c-MYC as a target of the APC pathway Science 281, 1509–1512 72 Plyte SE, Hughes K, Nikolakaki E, Pulverer BJ & Woodgett JR (1992) Glycogen synthase kinase-3: functions in oncogenesis and development Biochim Biophys Acta 1114, 147–162 73 Ross SE, Erickson RL, Hemati N & MacDougald OA (1999) Glycogen synthase kinase is an insulin-regulated C ⁄ EBPalpha kinase Mol Cell Biol 19, 8433– 8441 74 Cohen P (1999) The Croonian Lecture 1998 Identification of a protein kinase cascade of major importance in insulin signal transduction Philos Trans R Soc Lond B Biol Sci 354, 485–495 75 Cross DA, Alessi DR, Cohen P, Andjelkovich M & Hemmings BA (1995) Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B Nature 378, 785–789 76 Dajani R, Fraser E, Roe SM, Young N, Good V, Dale TC & Pearl LH (2001) Crystal structure of glycogen 5143 Flexible nets 77 78 79 80 81 82 83 84 85 86 87 88 89 90 synthase kinase beta: structural basis for phosphateprimed substrate specificity and autoinhibition Cell 105, 721–732 Hsu W, Zeng L & Costantini F (1999) Identification of a domain of Axin that binds to the serine ⁄ threonine protein phosphatase 2A and a self-binding domain J Biol Chem 274, 3439–3445 Seeling JM, Miller JR, Gil R, Moon RT, White R & Virshup DM (1999) Regulation of beta-catenin signaling by the B56 subunit of protein phosphatase 2A Science 283, 2089–2091 Grosschedl R, Giese K & Pagel J (1994) HMG domain proteins: architectural elements in the assembly of nucleoprotein structures Trends Genet 10, 94–100 Reeves R (2001) Molecular biology of HMGA proteins: hubs of nuclear function Gene 277, 63–81 Lehn DA, Elton TS, Johnson KR & Reeves R (1988) A conformational study of the sequence specific binding of HMG-I(Y) with the bovine interleukin-2 cDNA Biochem Int 16, 963–971 Evans JN, Nissen MS & R (1992) Assignment of the 1H NMR spectrum of a consensus DNA-binding peptide from the HMG-I protein Bull Mag Reson 14, 171–174 Evans JN, Zajicek J, Nissen MS, Munske G, Smith V & Reeves R (1995) 1H and 13C NMR assignments and molecular modelling of a minor groove DNAbinding peptide from the HMG-I protein Int J Pept Protein Res 45, 554–560 Lund T, Holtlund J, Fredriksen M & Laland SG (1983) On the presence of two new high mobility group-like proteins in HeLa S3 cells FEBS Lett 152, 163–167 Obradovic Z, Peng K, Vucetic S, Radivojac P & Dunker AK (2005) Exploiting heterogeneous sequence properties improves prediction of protein disorder Proteins: Struct Func Bioinf in press Reeves R & Nissen MS (1990) The A.T-DNA-binding domain of mammalian high mobility group I chromosomal proteins A novel peptide motif for recognizing DNA structure J Biol Chem 265, 8573–8582 Huth JR, Bewley CA, Nissen MS, Evans JN, Reeves R, Gronenborn AM & Clore GM (1997) The solution structure of an HMG-I(Y)-DNA complex defines a new architectural minor groove binding motif Nat Struct Biol 4, 657–665 Disney JE, Johnson KR, Magnuson NS, Sylvester SR & Reeves R (1989) High-mobility group protein HMG-I localizes to G ⁄ Q- and C-bands of human and mouse chromosomes J Cell Biol 109, 1975–1982 Reeves R (1992) Chromatin changes during the cell cycle Curr Opin Cell Biol 4, 413–423 Reeves R & Beckerbauer L (2001) HMGI ⁄ Y proteins: flexible regulators of transcription and chromatin structure Biochim Biophys Acta 1519, 13–29 5144 A K Dunker et al 91 Varshavsky A, Levinger L, Sundin O, Barsoum J, Ozkaynak E, Swerdlow P & Finley D (1983) Cellular and SV40 chromatin: replication, segregation, ubiquitination, nuclease-hypersensitive sites, HMGcontaining nucleosomes, and heterochromatin-specific protein Cold Spring Harb Symp Quant Biol 47 Part 1, 511–528 92 Reeves R & Nissen MS (1993) Interaction of high mobility group-I(Y) nonhistone proteins with nucleosome core particles J Biol Chem 268, 21137–21146 93 Chiappetta G, Avantaggiato V, Visconti R, Fedele M, Battista S, Trapasso F, Merciai BM, Fidanza V, Giancotti V, Santoro M, Simeone A & Fusco A (1996) High level expression of the HMGI(Y) gene during embryonic development Oncogene 13, 2439–2446 94 Bustin M & Reeves R (1996) High-mobility-group chromosomal proteins: architectural components that facilitate chromatin function Prog Nucleic Acid Res Mol Biol 54, 35–100 95 Friedberg EC, Walker GC & Siede W (1995) DNA Repair and Mutagenesis ASM Press, Washington, DC 96 Bootsma D, Kreamer KH, Cleaver JE & Hoeijmakers JHJ (1998) Nucleotide excision repair: xeroderma pigmentosum, Cocayne syndrome, and trichothiodystrophy In The Genetic Basis of Human Cancer (Vogelstein B & Kinzler KW, eds), pp 245–274 McGraw-Hill, New York 97 de Boer J & Hoeijmakers JH (2000) Nucleotide excision repair and human syndromes Carcinogenesis 21, 453–460 98 Cleaver JE & States JC (1997) The DNA damagerecognition problem in human and other eukaryotic cells: the XPA damage binding protein Biochem J 328, 1–12 99 Jones CJ & Wood RD (1993) Preferential binding of the xeroderma pigmentosum group A complementing protein to damaged DNA Biochemistry 32, 12096– 12104 100 Asahina H, Kuraoka I, Shirakawa M, Morita EH, Miura N, Miyamoto I, Ohtsuka E, Okada Y & Tanaka K (1994) The XPA protein is a zinc metalloprotein with an ability to recognize various kinds of DNA damage Mutat Res 315, 229–237 101 Nitta M, Saijo M, Kodo N, Matsuda T, Nakatsu Y, Tamai H & Tanaka K (2000) A novel cytoplasmic GTPase XAB1 interacts with DNA repair protein XPA Nucleic Acids Res 28, 4212–4218 102 Nakatsu Y, Asahina H, Citterio E, Rademakers S, Vermeulen W, Kamiuchi S, Yeo JP, Khaw MC, Saijo M, Kodo N, Matsuda T, Hoeijmakers JH & Tanaka K (2000) XAB2, a novel tetratricopeptide repeat protein involved in transcription-coupled DNA repair and transcription J Biol Chem 275, 34931–34937 103 Saijo M, Kuraoka I, Masutani C, Hanaoka F & Tanaka K (1996) Sequential binding of DNA repair FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al 104 105 106 107 108 109 110 111 112 113 114 115 proteins RPA and ERCC1 to XPA in vitro Nucleic Acids Res 24, 4719–4724 Li L, Lu X, Peterson CA & Legerski RJ (1995) An interaction between the DNA repair factor XPA and replication protein A appears essential for nucleotide excision repair Mol Cell Biol 15, 5396–5402 Miyamoto I, Miura N, Niwa H, Miyazaki J & Tanaka K (1992) Mutational analysis of the structure and function of the xeroderma pigmentosum group A complementing protein Identification of essential domains for nuclear localization and DNA excision repair J Biol Chem 267, 12182–12187 Nagai A, Saijo M, Kuraoka I, Matsuda T, Kodo N, Nakatsu Y, Mimaki T, Mino M, Biggerstaff M, Wood RD, Sijbers A, Hoeijmakers JHJ & Tanaka K (1995) Enhancement of damage-specific DNA binding of XPA by interaction with the ERCC1 DNA repair protein Biochem Biophys Res Commun 211, 960–966 Li L, Elledge SJ, Peterson CA, Bales ES & Legerski RJ (1994) Specific association between the human DNA repair proteins XPA and ERCC1 Proc Natl Acad Sci USA 91, 5012–5016 Park CH, Mu D, Reardon JT & Sancar A (1995) The general transcription-repair factor TFIIH is recruited to the excision repair complex by the XPA protein independent of the TFIIE transcription factor J Biol Chem 270, 4896–4902 Nocentini S, Coin F, Saijo M, Tanaka K & Egly JM (1997) DNA damage recognition by XPA protein promotes efficient recruitment of transcription factor II H J Biol Chem 272, 22991–22994 Kuraoka I, Morita EH, Saijo M, Matsuda T, Morikawa K, Shirakawa M & Tanaka K (1996) Identification of a damaged-DNA binding domain of the XPA protein Mutat Res 362, 87–95 Ikegami T, Kuraoka I, Saijo M, Kodo N, Kyogoku Y, Morikawa K, Tanaka K & Shirakawa M (1998) Solution structure of the DNA- and RPA-binding domain of the human repair factor XPA Nat Struct Biol 5, 701–706 Buchko GW, Ni S, Thrall BD & Kennedy MA (1998) Structural features of the minimal DNA binding domain (M98–F219) of human nucleotide excision repair protein XPA Nucleic Acids Res 26, 2779–2788 Iakoucheva LM, Kimzey AL, Masselon CD, Bruce JE, Garner EC, Brown CJ, Dunker AK, Smith RD & Ackerman EJ (2001) Identification of intrinsic order and disorder in the DNA repair protein XPA Protein Sci 10, 560–571 Fontana A, de Laureto PP, Spolaore B, Frare E, Picotti P & Zambonin M (2004) Probing protein structure by limited proteolysis Acta Biochim Pol 51, 299– 321 Fontana A, Polverino de Laureto P, De Filippis V, Scaramella E & Zambonin M (1997) Probing the FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS Flexible nets 116 117 118 119 120 121 122 123 124 125 126 127 128 partly folded states of proteins by limited proteolysis Fold Des 2, R17–R26 Morgan DO (1995) Principles of CDK regulation Nature 374, 131–134 Morgan DO (1997) Cyclin-dependent kinases: engines, clocks, and microprocessors Annu Rev Cell Dev Biol 13, 261–291 Nigg EA (2001) Mitotic kinases as regulators of cell division and its checkpoints Nat Rev Mol Cell Biol 2, 21–32 Pavletich NP (1999) Mechanisms of cyclin-dependent kinase regulation: structures of Cdks, their cyclin activators, and Cip and INK4 inhibitors J Mol Biol 287, 821–828 Russo AA, Jeffrey PD, Patten AK, Massague J & Pavletich NP (1996) Crystal structure of the p27Kip1 cyclin-dependent-kinase inhibitor bound to the cyclin A-Cdk2 complex Nature 382, 325–331 Byth KF, Cooper N, Culshaw JD, Heaton DW, Oakes SE, Minshull CA, Norman RA, Pauptit RA, Tucker JA, Breed J, Pannifer A, Rowsell S, Stanway JJ, Valentine AL & Thomas AP (2004) Imidazo[1,2-b]pyridazines: a potent and selective class of cyclin-dependent kinase inhibitors Bioorg Med Chem Lett 14, 2249– 2252 Lacy ER, Filippov I, Lewis WS, Otieno S, Xiao L, Weiss S, Hengst L & Kriwacki RW (2004) p27 binds cyclin-CDK complexes through a sequential mechanism involving binding-induced protein folding Nat Struct Mol Biol 11, 358–364 Harper JW, Adami GR, Wei N, Keyomarsi K & Elledge SJ (1993) The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases Cell 75, 805–816 Harper JW, Elledge SJ, Keyomarsi K, Dynlacht B, Tsai LH, Zhang P, Dobrowolski S, Bai C, ConnellCrowley L, Swindell E, Fox MP & Wei N (1995) Inhibition of cyclin-dependent kinases by p21 Mol Biol Cell 6, 387–400 Polyak K, Lee MH, Erdjument-Bromage H, Koff A, Roberts JM, Tempst P & Massague J (1994) Cloning of p27Kip1, a cyclin-dependent kinase inhibitor and a potential mediator of extracellular antimitogenic signals Cell 78, 59–66 Toyoshima H & Hunter T (1994) p27, a novel inhibitor of G1 cyclin-Cdk protein kinase activity, is related to p21 Cell 78, 67–74 Matsuoka S, Edwards MC, Bai C, Parker S, Zhang P, Baldini A, Harper JW & Elledge SJ (1995) p57KIP2, a structurally distinct member of the p21CIP1 Cdk inhibitor family, is a candidate tumor suppressor gene Genes Dev 9, 650–662 Flaugh SL & Lumb KJ (2001) Effects of macromolecular crowding on the intrinsically disordered proteins c-Fos and p27(Kip1) Biomacromolecules 2, 538–540 5145 Flexible nets 129 Bienkiewicz EA, Adkins JN & Lumb KJ (2002) Functional consequences of preorganized helical structure in the intrinsically disordered cell-cycle inhibitor p27(Kip1) Biochemistry 41, 752–759 130 Spolar RS & Record MT Jr (1994) Coupling of local folding to site-specific binding of proteins to DNA Science 263, 777–784 131 Cohen P & Klee CB (1988) Calmodulin Elsevier, New York 132 Vetter SW & Leclerc E (2003) Novel aspects of calmodulin target recognition and activation Eur J Biochem 270, 404–414 133 Yap KL, Kim J, Truong K, Sherman M, Yuan T & Ikura M (2000) Calmodulin target database J Struct Funct Genomics 1, 8–14 134 Rhoads AR & Friedberg F (1997) Sequence motifs for calmodulin recognition FASEB J 11, 331–340 135 Babu YS, Bugg CE & Cook WJ (1988) Structure of ˚ calmodulin refined at 2.2 A resolution J Mol Biol 204, 191–204 136 Barbato G, Ikura M, Kay LE, Pastor RW & Bax A (1992) Backbone dynamics of calmodulin studied by 15N relaxation using inverse detected two-dimensional NMR spectroscopy: the central helix is flexible Biochemistry 31, 5269–5278 137 Fernandez A (2004) Functionality of wrapping defects in soluble proteins: what cannot be kept dry must be conserved J Mol Biol 337, 477–483 138 Fernandez A, Colubri A & Berry RS (2002) Threebody correlations in protein folding: the origin of cooperativity Physica A 307, 235–259 139 Fernandez A & Scheraga HA (2003) Insufficiently dehydrated hydrogen bonds as determinants of protein interactions Proc Natl Acad Sci USA 100, 113–118 140 Fernandez A & Scott LR (2003) Adherence of packing defects in soluble proteins Phys Rev Lett 91, 018102 141 Fernandez A & Scott R (2003) Dehydron: a structurally encoded signal for protein interaction Biophys J 85, 1914–1928 142 Fernandez A, Scott R & Berry RS (2004) The nonconserved wrapping of conserved protein folds reveals a trend toward increasing connectivity in proteomic networks Proc Natl Acad Sci USA 101, 2823–2827 143 Qin H, Lu HH, Wu WB & Li WH (2003) Evolution of the yeast protein interaction network Proc Natl Acad Sci USA 100, 12820–12824 144 Fernandez A & Berry RS (2004) Molecular dimension explored in evolution to promote proteomic complexity Proc Natl Acad Sci USA 101, 13460–13465 145 Bourhis JM, Johansson K, Receveur-Brechot V, Oldfield CJ, Dunker KA, Canard B & Longhi S (2004) The C-terminal domain of measles virus nucleoprotein belongs to the class of intrinsically disordered proteins that fold upon binding to their physiological partner Virus Res 99, 157–167 5146 A K Dunker et al 146 Oldfield CJ (2002) Predictions of disordered proteins and protein binding regions: applications to structural genomics Honors Thesis, Washington State University Pullman, Washington 147 Oldfield CJ, Cheng Y, Cortese MS, Romero P, Uversky VN & Dunker AK (2005) Coupled folding and binding with a-helix-forming molecular recognition elements Biochemistry 44, 12454–12470 148 Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, Shindyalov IN & Bourne PE (2000) The Protein Data Bank Nucleic Acids Res 28, 235–542 149 Garner E, Romero P, Dunker AK, Brown C & Obradovic Z (1999) Predicting Binding Regions within Disordered Proteins Genome Inform Series Workshop Genome Inform 10, 41–50 150 Callaghan AJ, Aurikko JP, Ilag LL, Gunter Grossmann J, Chandran V, Kuhnel K, Poljak L, Carpousis AJ, Robinson CV, Symmons MF & Luisi BF (2004) Studies of the RNA degradosome-organizing domain of the Escherichia coli ribonuclease RNase E J Mol Biol 340, 965–979 151 Wagner A (2001) The yeast protein interaction network evolves rapidly and contains few redundant duplicate genes Mol Biol Evol 18, 1283–1292 152 Ideker T, Ozier O, Schwikowski B & Siegel AF (2002) Discovering regulatory and signalling circuits in molecular interaction networks Bioinformatics 18 (Suppl 1), S233–S240 153 Galitski T (2004) Molecular networks in model systems Annu Rev Genomics Hum Genet 5, 177–187 154 Weinreb PH, Zhen W, Poon AW, Conway KA & Lansbury PT Jr (1996) NACP, a protein implicated in Alzheimer’s disease and learning, is natively unfolded Biochemistry 35, 13709–13715 155 Dev KK, van der Putten H, Sommer B & Rovelli G (2003) Part I: parkin-associated proteins and Parkinson’s disease Neuropharmacology 45, 1–13 156 Dev KK, Hofele K, Barbieri S, Buchman VL & van der Putten H (2003) Part II: alpha-synuclein and its molecular pathophysiological role in neurodegenerative disease Neuropharmacology 45, 14–44 157 Lynch WP, Riseman VM & Bretscher A (1987) Smooth muscle caldesmon is an extended flexible monomeric protein in solution that can readily undergo reversible intra- and intermolecular sulfhydryl cross-linking A mechanism for caldesmon’s F-actin bundling activity J Biol Chem 262, 7429–7437 158 Gerthoffer WT (2005) Signal-transduction pathways that regulate visceral smooth muscle function III Coupling of muscarinic receptors to signaling kinases and effector proteins in gastrointestinal smooth muscles Am J Physiol Gastrointest Liver Physiol 288, G849–G853 159 Polyakov AA, Huber PA, Marston SB & Gusev NB (1998) Interaction of isoforms of S100 protein with smooth muscle caldesmon FEBS Lett 422, 235–239 FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS A K Dunker et al 160 Huber PA (1997) Caldesmon Int J Biochem Cell Biol 29, 1047–1051 161 Reeves R & Nissen MS (1999) Purification and assays for high mobility group HMG-I(Y) protein function Methods Enzymol 304, 155–188 162 Sgarra R, Tessari MA, Di Bernardo J, Rustighi A, Zago P, Liberatori S, Armini A, Bini L, Giancotti V & Manfioletti G (2005) Discovering high mobility group A molecular partners in tumour cells Proteomics 5, 1494–1506 163 Fasshauer D, Otto H, Eliason WK, Jahn R & Brunger AT (1997) Structural changes are associated with soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor complex formation J Biol Chem 272, 28036–28041 164 Perez-Branguli F, Muhaisen A & Blasi J (2002) Munc 18a binding to syntaxin 1A and 1B isoforms defines its localization at the plasma membrane and blocks SNARE assembly in a three-hybrid system assay Mol Cell Neurosci 20, 169–180 165 Annaert WG, Becker B, Kistner U, Reth M & Jahn R (1997) Export of cellubrevin from the endoplasmic reticulum is controlled by BAP31 J Cell Biol 139, 1397– 1410 166 Weir ML, Klip A & Trimble WS (1998) Identification of a human homologue of the vesicle-associated membrane protein (VAMP)-associated protein of 33 kDa (VAP-33): a broadly expressed protein that binds to VAMP Biochem J 333, 247–251 167 Nishimura Y, Hayashi M, Inada H & Tanaka T (1999) Molecular cloning and characterization of mammalian homologues of vesicle-associated membrane protein-associated (VAMP-associated) proteins Biochem Biophys Res Commun 254, 21–26 168 Ravichandran V, Chawla A & Roche PA (1996) Identification of a novel syntaxin- and synaptobrevin ⁄ VAMP-binding protein, SNAP-23, expressed in nonneuronal tissues J Biol Chem 271, 13300–13303 169 Mark WY, Liao JC, Lu Y, Ayed A, Laister R, Szymczyna B, Chakrabartty A & Arrowsmith CH (2005) Characterization of segments from the central region of BRCA1: an intrinsically disordered scaffold for multiple protein–protein and protein–DNA interactions? J Mol Biol 345, 275–287 170 Yoshida K & Miki Y (2004) Role of BRCA1 and BRCA2 as regulators of DNA repair, transcription, and cell cycle in response to DNA damage Cancer Sci 95, 866–871 171 Saijo M, Matsuda T, Kuraoka I & Tanaka K (2004) Inhibition of nucleotide excision repair by anti-XPA monoclonal antibodies which interfere with binding to RPA, ERCC1, and TFIIH Biochem Biophys Res Commun 321, 815–822 172 Warnmark A, Wikstrom A, Wright AP, Gustafsson JA & Hard T (2001) The N-terminal regions of FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS Flexible nets 173 174 175 176 177 178 179 180 181 182 183 184 185 estrogen receptor alpha and beta are unstructured in vitro and show different TBP binding properties J Biol Chem 276, 45939–45944 Liu G, Schwartz JA & Brooks SC (2000) Estrogen receptor protects p53 from deactivation by human double minute-2 Cancer Res 60, 1810–1814 Fan S, Wang J, Yuan R, Ma Y, Meng Q, Erdos MR, Pestell RG, Yuan F, Auborn KJ, Goldberg ID & Rosen EM (1999) BRCA1 inhibition of estrogen receptor signaling in transfected cells Science 284, 1354– 1356 Garcia Pedrero JM, Del Rio B, Martinez-Campa C, Muramatsu M, Lazo PS & Ramos S (2002) Calmodulin is a selective modulator of estrogen receptors Mol Endocrinol 16, 947–960 Chang J, Kim DH, Lee SW, Choi KY & Sung YC (1995) Transactivation ability of p53 transcriptional activation domain is directly related to the binding affinity to TATA-binding protein J Biol Chem 270, 25014–25019 Derbyshire DJ, Basu BP, Serpell LC, Joo WS, Date T, Iwabuchi K & Doherty AJ (2002) Crystal structure of human 53BP1 BRCT domains bound to p53 tumour suppressor EMBO J 21, 3863–3872 Joerger AC, Allen MD & Fersht AR (2004) Crystal structure of a superstable mutant of human p53 core domain Insights into the mechanism of rescuing oncogenic mutations J Biol Chem 279, 1291–1296 Kuszewski J, Gronenborn AM & Clore GM (1999) Improving the packing and accuracy of NMR structures with a pseudopotential for the radius of gyration J Am Chem Soc 121, 2337–2338 Sengupta S & Harris CC (2005) p53: traffic cop at the crossroads of DNA repair and recombination Nat Rev Mol Cell Biol 6, 44–55 Uhrinova S, Uhrin D, Powers H, Watt K, Zheleva D, Fischer P, McInnes C & Barlow PN (2005) Structure of free MDM2 N-terminal domain reveals conformational adjustments that accompany p53-binding J Mol Biol 350, 587–598 Kussie PH, Gorina S, Marechal V, Elenbaas B, Moreau J, Levine AJ & Pavletich NP (1996) Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain Science 274, 948–953 Bothner B, Lewis WS, DiGiammarino EL, Weber JD, Bothner SJ & Kriwacki RW (2001) Defining the molecular basis of Arf and Hdm2 interactions J Mol Biol 314, 263–277 Iwakuma T & Lozano G (2003) MDM2, an introduction Mol Cancer Res 1, 993–1000 Kissinger CR, Parge HE, Knighton DR, Lewis CT, Pelletier LA, Tempczyk A, Kalish VJ, Tucker KD, Showalter RE, Moomaw EW, Gastinel LN, Habuka N, Chen X, Maldonado F, Barker JE, Bacquet R & Villafranca JE (1995) Crystal structures of human 5147 Flexible nets 186 187 188 189 190 191 192 193 194 calcineurin and the human FKBP12-FK506-calcineurin complex Nature 378, 641–644 Im SH & Rao A (2004) Activation and deactivation of gene expression by Ca2+ ⁄ calcineurin-NFAT-mediated signaling Mol Cells 18, 1–9 Genesca L, Aubareda A, Fuentes JJ, Estivill X, De La Luna S & Perez-Riba M (2003) Phosphorylation of calcipressin increases its ability to inhibit calcineurin and decreases calcipressin half-life Biochem J 374, 567–575 Sun L, Youn HD, Loh C, Stolow M, He W & Liu JO (1998) Cabin 1, a negative regulator for calcineurin signaling in T lymphocytes Immunity 8, 703–711 Banerjee A, Banks AS, Nawijn MC, Chen XP & Rothman PB (2002) Cutting edge: Suppressor of cytokine signaling inhibits activation of NFATp J Immunol 168, 4277–4281 Frey N, Richardson JA & Olson EN (2000) Calsarcins, a novel family of sarcomeric calcineurin-binding proteins Proc Natl Acad Sci USA 97, 14632–14637 Rittinger K, Budman J, Xu J, Volinia S, Cantley LC, Smerdon SJ, Gamblin SJ & Yaffe MB (1999) Structural analysis of 14-3-3 phosphopeptide complexes identifies a dual role for the nuclear export signal of 14-3-3 in ligand binding Mol Cell 4, 153–166 Fu H, Subramanian RR & Masters SC (2000) 14-3-3 proteins: structure, function, and regulation Annu Rev Pharmacol Toxicol 40, 617–647 Dehde S, Rohaly G, Schub O, Nasheuer HP, Bohn W, Chemnitz J, Deppert W & Dornreiter I (2001) Two immunologically distinct human DNA polymerase alpha-primase subpopulations are involved in cellular DNA replication Mol Cell Biol 21, 2581–2593 Ruffner H, Jiang W, Craig AG, Hunter T & Verma IM (1999) BRCA1 is phosphorylated at serine 1497 in vivo at a cyclin-dependent kinase phosphorylation site Mol Cell Biol 19, 4843–4854 5148 A K Dunker et al 195 Morton WM, Ayscough KR & McLaughlin PJ (2000) Latrunculin alters the actin–monomer subunit interface to prevent polymerization Nat Cell Biol 2, 376–378 196 Schutt CE, Myslik JC, Rozycki MD, Goonesekere NC & Lindberg U (1993) The structure of crystalline profilin-beta-actin Nature 365, 810–816 197 Ballweber E, Galla M, Aktories K, Yeoh S, Weeds AG & Mannherz HG (2001) Interaction of ADP-ribosylated actin with actin binding proteins FEBS Lett 508, 131–135 198 Xia Z & Storm DR (2005) The role of calmodulin as a signal integrator for synaptic plasticity Nat Rev Neurosci 6, 267–276 199 Takahashi K, Hiwada K & Kokubu T (1988) Vascular smooth muscle calponin A novel troponin T-like protein Hypertension 11, 620–626 200 Gusev NB (2001) Some properties of caldesmon and calponin and the participation of these proteins in regulation of smooth muscle contraction and cytoskeleton formation Biochemistry (Mosc) 66, 1112–1121 201 Bader GD, Betel D & Hogue CW (2003) BIND: the Biomolecular Interaction Network Database Nucleic Acids Res 31, 248–250 202 Salwinski L, Miller CS, Smith AJ, Pettit FK, Bowie JU & Eisenberg D (2004) The Database of Interacting Proteins: 2004 update Nucleic Acids Res 32, D449–D451 203 von Mering C, Jensen LJ, Snel B, Hooper SD, Krupp M, Foglierini M, Jouffre N, Huynen MA & Bork P (2005) STRING: known and predicted protein–protein associations, integrated and transferred across organisms Nucleic Acids Res 33, D433–D437 204 Peri S, Navarro JD, Kristiansen TZ, Amanchy R, Surendranath V, Muthusamy B, Gandhi TK, Chandrika KN, Deshpande N, Suresh S et al (2004) Human protein reference database as a discovery resource for proteomics Nucleic Acids Res 32, D497–D501 205 Barabasi AL (2001) The physics of the Web Physics World 14, 33–38 FEBS Journal 272 (2005) 5129–5148 ª 2005 FEBS ... number of protein? ? ?protein interactions ⁄ number of complexes (inter ⁄ comp.), while the STRING search results are presented as number of protein? ? ?protein interactions (inter.) only Proteins are... contained regions of intrinsic disorder One example of an ordered hub, calmodulin, makes use of a flexible hinge to facilitate binding diversity Each of these classes and their roles in protein? ? ?protein. .. important roles in the binding interactions of these hub proteins The xeroderma pigmentosum group A protein (XPA) represents an interesting example of a partially disordered hub protein Xeroderma

Ngày đăng: 07/03/2014, 21:20

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan