2008 solution structure of the c-terminal dimerization domain of sars coronavirus nucleocapsid...

15
Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAIL-NMR Method Mitsuhiro Takeda 1 , Chung-ke Chang 2 , Teppei Ikeya 1,3 , Peter Güntert 3,4 , Yuan-hsiang Chang 2 , Yen-lan Hsu 2 , Tai-huang Huang 2,5,6 and Masatsune Kainosho 1,3 1 Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan 2 Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, ROC 3 Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-ohsawa, Hachioji 192-0397, Japan 4 Institute of Biophysical Chemistry, J.W. Goethe- University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany 5 Department of Physics, National Taiwan Normal University, Taipei, Taiwan, ROC 6 The Genomic Research Center, Academia Sinica, Taipei, Taiwan, ROC Received 26 October 2007; received in revised form 24 November 2007; accepted 28 November 2007 Available online 5 December 2007 The C-terminal domain (CTD) of the severe acute respiratory syndrome coronavirus (SARS-CoV) nucleocapsid protein (NP) contains a potential RNA-binding region in its N-terminal portion and also serves as a dimerization domain by forming a homodimer with a molecular mass of 28 kDa. So far, the structure determination of the SARS-CoV NP CTD in solution has been impeded by the poor quality of NMR spectra, especially for aromatic resonances. We have recently developed the stereo-array isotope labeling (SAIL) method to overcome the size problem of NMR structure determination by utilizing a protein exclusively composed of stereo- and regio-specifically isotope-labeled amino acids. Here, we employed the SAIL method to determine the high-quality solution structure of the SARS-CoV NP CTD by NMR. The SAIL protein yielded less crowded and better resolved spectra than uniform 13 C and 15 N labeling, and enabled the homodimeric solution structure of this protein to be determined. The NMR structure is almost identical with the previously solved crystal structure, except for a disordered putative RNA-binding domain at the N-terminus. Studies of the chemical shift perturbations caused by the binding of single-stranded DNA and mutational analyses have identified the disordered region at the N-termini as the prime site for nucleic acid binding. In addition, residues in the β-sheet region also showed significant perturbations. Mapping of the locations of these residues onto the helical model observed in the crystal revealed that these two regions are parts of the interior lining of the positively charged helical groove, supporting the hypothesis that the helical oligomer may form in solution. © 2008 Published by Elsevier Ltd. Edited by M. F. Summers Keywords: SAIL-NMR; SARS nucleocapsid protein; protein structure determination; nucleocapsid packaging *Corresponding authors. T. Huang is to be contacted at Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, ROC. M. Kainosho, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan. E-mail addresses: [email protected]; [email protected]. M.T. and C.C. contributed equally to this work. Abbreviations used: CTD, C-terminal domain containing residues 248365; SARS-CoV, severe acute respiratory syndrome coronavirus; NP, nucleocapsid protein; SAIL, stereo-array isotope labeling; NTD, N-terminal domain containing residues 45181; NOE, nuclear Overhauser effect; EMSA, electrophoretic mobility shift assay; HSQC, heteronuclear single-quantum coherence; CT, constant time; UL, uniformly labeled; PDB, Protein Data Bank; CSD, chemical shift displacement; poly-dT, poly-deoxythymine; ssDNA, single-stranded DNA; EDTA, ethylenediaminetetraacetic acid. doi:10.1016/j.jmb.2007.11.093 J. Mol. Biol. (2008) 380, 608622 Available online at www.sciencedirect.com 0022-2836/$ - see front matter © 2008 Published by Elsevier Ltd.

Upload: others

Post on 11-Sep-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

doi:10.1016/j.jmb.2007.11.093 J. Mol. Biol. (2008) 380, 608–622

Available online at www.sciencedirect.com

Solution Structure of the C-terminal DimerizationDomain of SARS Coronavirus Nucleocapsid ProteinSolved by the SAIL-NMR Method

Mitsuhiro Takeda1†, Chung-ke Chang2†, Teppei Ikeya1,3,Peter Güntert3,4, Yuan-hsiang Chang2, Yen-lan Hsu2,Tai-huang Huang2,5,6⁎ and Masatsune Kainosho1,3⁎

1Graduate School of Science,Nagoya University, Furo-cho,Chikusa-ku, Nagoya 464-8602,Japan2Institute of BiomedicalSciences, Academia Sinica,Taipei 115, Taiwan, ROC3Graduate School of Science,Tokyo Metropolitan University,1-1 Minami-ohsawa,Hachioji 192-0397, Japan4Institute of BiophysicalChemistry, J.W. Goethe-University Frankfurt am Main,Max-von-Laue-Str. 9, 60438Frankfurt am Main, Germany5Department of Physics, NationalTaiwan Normal University,Taipei, Taiwan, ROC6The Genomic Research Center,Academia Sinica, Taipei, Taiwan,ROC

Received 26 October 2007;received in revised form24 November 2007;accepted 28 November 2007Available online5 December 2007

*Corresponding authors. T. Huang iTaiwan, ROC. M. Kainosho, GraduaJapan. E-mail addresses: bmthh@ibm† M.T. and C.C. contributed equaAbbreviations used: CTD, C-term

syndrome coronavirus; NP, nucleoccontaining residues 45–181; NOE, nuheteronuclear single-quantumcohereshift displacement; poly-dT, poly-de

0022-2836/$ - see front matter © 2008 P

The C-terminal domain (CTD) of the severe acute respiratory syndromecoronavirus (SARS-CoV) nucleocapsid protein (NP) contains a potentialRNA-binding region in its N-terminal portion and also serves as adimerization domain by forming a homodimer with a molecular mass of28 kDa. So far, the structure determination of the SARS-CoV NP CTD insolution has been impeded by the poor quality of NMR spectra, especiallyfor aromatic resonances. We have recently developed the stereo-arrayisotope labeling (SAIL) method to overcome the size problem of NMRstructure determination by utilizing a protein exclusively composed ofstereo- and regio-specifically isotope-labeled amino acids. Here, weemployed the SAIL method to determine the high-quality solution structureof the SARS-CoV NP CTD by NMR. The SAIL protein yielded less crowdedand better resolved spectra than uniform 13C and 15N labeling, and enabledthe homodimeric solution structure of this protein to be determined. TheNMR structure is almost identical with the previously solved crystalstructure, except for a disordered putative RNA-binding domain at theN-terminus. Studies of the chemical shift perturbations caused by thebinding of single-stranded DNA and mutational analyses have identifiedthe disordered region at the N-termini as the prime site for nucleic acidbinding. In addition, residues in the β-sheet region also showed significantperturbations. Mapping of the locations of these residues onto the helicalmodel observed in the crystal revealed that these two regions are parts ofthe interior lining of the positively charged helical groove, supporting thehypothesis that the helical oligomer may form in solution.

© 2008 Published by Elsevier Ltd.

Keywords: SAIL-NMR; SARS nucleocapsid protein; protein structuredetermination; nucleocapsid packaging

Edited by M. F. Summers

s to be contacted at Institute of Biomedical Sciences, Academia Sinica, Taipei 115,te School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602,s.sinica.edu.tw; [email protected].

lly to this work.inal domain containing residues 248–365; SARS-CoV, severe acute respiratoryapsid protein; SAIL, stereo-array isotope labeling; NTD, N-terminal domainclear Overhauser effect; EMSA, electrophoretic mobility shift assay; HSQC,nce; CT, constant time;UL, uniformly labeled; PDB, ProteinData Bank;CSD, chemicaloxythymine; ssDNA, single-stranded DNA; EDTA, ethylenediaminetetraacetic acid.

ublished by Elsevier Ltd.

Page 2: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

609Structure of SAIL SARS-CoV N Protein

Introduction

Severe acute respiratory syndrome (SARS) is arecently emergent disease caused by the SARS-associated coronavirus (CoV).1,2 The SARS-CoVnucleocapsid protein (NP) packages the viral geno-mic RNA into a ribonucleoprotein complex and iscrucial for the assembly of infectious virus particles.Based on comparative NMR studies of SARS-CoVNP deletion constructs, the protein contains twostructural domains: the N-terminal domain (NTD;containing residues 45–181) and the C-terminaldomain (CTD; containing residues 248–365),3

flanked by the long, disordered N- and C-terminiand the linker sequence (Fig. 1a). The NTDreportedly acts as a putative RNA-binding domain,and the CTD functions as a dimerization domain.3,4

Recently, the CTD has also been shown to bindnucleic acids with high affinity.5 Structural investi-gations of the isolated SARS-CoV NP CTD havebeen performed by NMR and X-ray crystallography.In a previous NMR study, the topological structureof the isolated SARS-CoV NP CTD as a homodimerwas elucidated based on limited intersubunit andintrasubunit nuclear Overhauser effects (NOEs).6

This topology was confirmed by the subsequentlyreported crystal structures of the SARS-CoV NPCTD and of a shorter construct spanning residues270–370.5,7 The crystal structure of the CTDrevealed that residues 248–280 form a positivelycharged patch, which acts as a putative oligonucleo-tide-binding region. The patch also participates inintermolecular and intramolecular interactionswithin the crystal, resulting in the formation of anoctameric asymmetric unit. However, previous bio-chemical and biophysical studies have shown thatthe CTD exists solely as a dimer in solution.6 Thesefindings motivated us to investigate the nature ofthe CTD in solution. Initial attempts at the completestructure elucidation of the SARS-CoV NP CTDthrough NMR were impeded by its short T2 relaxa-tion times and significant peak overlaps.6

Recently, we have developed the stereo-arrayisotope labeling (SAIL) method, which utilizes pro-teins exclusively composed of stereo- and regio-specifically labeled amino acids.8 Compared to

conventional uniform 13C,15N isotopic labeling, thequality of the spectra of the SAIL sample wassufficiently improved, so that a high-resolutionsolution structure determination could be per-formed. The sharpened resonance lines and reducedpeak overlap with the SAIL method are due to theselective deuteration of many nonlabile protons. Theremaining protons with known stereo-specificassignments provide plentiful information aboutthe structure of the protein. In this study, we per-formed an NMR study of the SARS-CoV NP CTDwith the help of the SAIL method. Compared to theprotein uniformly labeled with 13C and 15N, theSAIL sample significantly improved the quality ofthe NMR spectra, to the extent that a high-resolutionstructure determination could be performed. Thetertiary structure obtained by NMR is almost iden-tical with that of the crystal structure, except fora disordered putative RNA-binding domain at theN-terminus. We further applied NMR, mutationanalyses, and electrophoretic mobility shift assays(EMSAs) to pinpoint the nucleic-acid-binding site.The active site thus identified agrees well with thehelical ribonucleoprotein model suggested by thecrystal structure.

Results

Preparation of the SAIL sample of the SARS-CoVNP CTD

The preparation of proteins composed of SAILamino acids requires cell-free expression to effi-ciently incorporate the SAIL amino acids into theprotein without them being affected by metabolicscrambling in living cells. The expression of theSARS-CoV NP CTD was initially examined in asmall-scale reaction (Fig. 1b). Subsequently, the1H–15N heteronuclear single-quantum coherence(HSQC) of the 15N-labeled SARS-CoV NP CTDproduced by cell-free expression was comparedwith that produced by in vivo expression. Thesespectra were identical, thus confirming that thestructures of the proteins produced by cell-free andin vivo expressions are identical (data not shown).

Fig. 1. Preparation of the SARS-CoV NP CTD. (a) Schematic dia-gram of the domain architecture ofthe SARS-CoVNP. (b) SDS-PAGE ofthe cell-free reaction mixture for theSARS-CoV NP CTD. The left laneshows the molecular weight mar-kers. The band corresponding to themonomer of the SARS-CoV NPCTD is labeled with an arrow.

Page 3: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

610 Structure of SAIL SARS-CoV N Protein

The final sample of the SAIL SARS-CoV NP CTDwas produced by using the Escherichia coli cell-freeprotein synthesis system, with optimizations for theproduction of labeled NMR samples.9

The SAIL method improves the quality of theNMR spectrum

The 1H–13C constant time (CT) HSQC spectra ofan aliphatic region were compared between uni-formly labeled (UL) and SAIL samples under thesame conditions. In the case of the UL sample, thesignals were prone to overlap between diastereo-topic pairs, and some signals were severely broa-dened beyond detection in the methylene region(Fig. 2a). In contrast, the corresponding spectrumfrom the SAIL sample had much better quality thanthat from the UL sample (Fig. 2b). The signal/noiseratios for the SAIL sample were several times higherthan those for the corresponding UL sample, con-sistent with previous results for calmodulin- and

Fig. 2. Comparisons of NMR spectra for the SARS-CoV NAliphatic region of 1H–13C CT-HSQC for UL (a) and SAIL (b) Ssame conditions. The sample concentration was 0.5 mM. In (bsections from (a) (red) and (b) (black). The peak scales are ide

maltodextrin-binding proteins (Fig. 2c).8 For theSARS-CoV NP CTD, the number of peaks to beobserved theoretically in the 1H–13C CT HSQCspectra, including methyl, methylene, and methaneprotons, decreased from 517 for the UL sample to343 for the SAIL sample, greatly simplifying theanalytical process.For the aromatic region, improvement due to the

use of the SAIL method was even more striking. Thearomatic rings of UL Phe and Tyr contain four andfive 13C–1H pairs, respectively (Fig. 3a). In the caseof SAIL, the six-membered aromatic rings arelabeled by alternating 13C–1H and 12C–2H moieties(13C at the ε and γ positions; 12C at the δ and ζpositions) (Fig. 3b).10 In the UL sample, signals forthe 1H–13C moieties at the δ, ε, and ζ positions ofPhe, and at the δ position of Tyr, are overlappedaround 131 ppm in the carbon dimension, thusresulting in severe spectral crowding (Fig. 3c). Incontrast, the corresponding region for the SAILsample was much simpler due to the presence of

P CTD between UL and SAIL in the methylene region.ARS-CoV NP CTD. Both spectra were acquired under the), assignments for the SAIL sample are labeled. (c) Cross-ntical between the UL and SAIL spectra.

Page 4: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Fig. 3. Comparisons of NMR spectra for the SARS-CoV NP CTD between UL and SAIL in the aromatic region.Chemical structures of the aromatic rings for UL (a) and SAIL (c) phenylalanine. (b and d) Phenylalanine signals of 1H–13CHSQC for UL (b) and SAIL (d) SARS-CoV NP CTD. (e and f) Tyrosine signals of 1H–13C HSQC for UL (e) and SAIL (f)SARS-CoV NP CTD. To demonstrate the absence of the 1Jcc coupling of aromatic rings for SAIL phenylalanine andtyrosine residues, all 1H–13C HSQC spectra for the aromatic regions were recorded without the CT technique.

611Structure of SAIL SARS-CoV N Protein

signals exclusively from the 1H–13C moieties at theε position of Phe. The short relaxation times ledto the detection of resonances that were severelybroadened beyond detection with the UL sample(Fig. 3d). Since one-bond 13C–13C couplings in theUL protein did not exist in the SAIL protein, asshown in the Tyr Hε–Cε region (Fig. 3e and f), theCT technique with a long evolution time (∼17 ms)is thus not required for the SAIL sample.10

With the benefits mentioned above, we were ableto acquire a set of NMR spectra with high sensitivity,and the expected chemical shifts were assigned to91.2% completeness for the SAIL sample.

Solution structure of SARS-CoV NP CTD

Even though the SARS-CoV NP CTD existed as anoctamer in the asymmetric unit in the crystal, abun-dant evidence suggests that it exists as a homodimerin solution.3,6 We manually assigned some of theintersubunit NOE peaks from the SARS-CoV NPCTD based on previous NMR work.6 These manu-ally assigned intersubunit distance restraints were

included in the combined automated assignments ofthe NOE peaks and the structure calculation byCYANA.11 NOE-derived distance restraints totalling2615 were obtained. Out of 100 structures calcu-lated, the 20 structures with the lowest targetfunction values were selected and energy-refinedas the final structures of the SARS-CoV NP CTD.The structural statistics for the NMR structure aresummarized in Table 1. Consistent with the pre-viously reported topological structure of the SARS-CoV NP CTD, the NMR structure of the SARS-CoVNP CTD adopts a domain-swapped homodimerconformation (Fig. 4a and b). The resulting struc-tures exhibited good convergence for both the back-bone and the side chains when the regions betweenresidues 260 and 365 of each dimer were super-imposed (Fig. 4a). The model has a disorderedregion spanning residues 248–259 and protrudingfrom the dimer core, which appears to be the resultof internal dynamics and prevents the detection oflong-range NOEs based on heteronuclear NOEmea-surements. 15N–{1H} NOE experiments recordedfor the SARS-CoV NP CTD showed that the hetero-

Page 5: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Table 1. Structural statistics for the NMR structure ofSARS-CoV CTD

Parameter Quantity

Completeness of chemical shift assignments (%) 91.2Total NOE upper distance bound restraints 2615

Short range (|i− j|≤1) 1313Medium range (1b|i− j|b5) 586Long range (|i− j|≤5) 716Intermolecular 260

Dihedral angle restraints (φ and ψ) 236CYANA target function (Å2) 2.55AMBER energy (kcal/mol) −6106Ramachandran plot statistics (%)

Most favored regions 84.6Additionally allowed regions 14.8Generously allowed regions 0.6Disallowed regions 0.0

Backbone RMSD for residues 260–365 (Å) 0.77All heavy atom RMSD for residues 260–365 (Å) 1.19

612 Structure of SAIL SARS-CoV N Protein

nuclear NOE values for this region are smaller thanthose for the structured region, indicating that thetwo N-termini are flexible in solution (data notshown). The secondary structural elements of theCTD in solution were defined based on the DSSPalgorithm,12 and they corresponded well to thoseidentified in the previousNMRstudy (Fig. 4b and c).6

Prior to our NMR investigation, the crystal struc-tures of constructs spanning residues 270–370[Protein Data Bank (PDB) ID 2gib] and residues248–365 (PDB ID 2cjr) were elucidated.5,7 Theoverall folds and secondary structure arrangementsare very similar between the crystal structures andthe NMR structure (Fig. 4a and c). The backboneRMSD between the protomers of the mean NMRstructure and the crystal structure of the CTDspanning residues 248–365 is 1.45 Å if residues260–319 and 333–358 are superimposed. If the NMRstructure is superimposed with the crystal structurespanning residues 270–370, then the backboneRMSD between the protomers is 1.26 Å for theregions of residues 274–319 and 333–358. There aretwo differences, however: first, in both crystal struc-tures, the β-sheet is distorted around residues 320–332 compared to those of the NMR structure (upperleft of Fig. 5a and b). Second, the two N-termini(residues 248–259) protruding from the dimer coreare disordered in the NMR structure, whereas, in thecrystal structure, they are involved in a number ofintramonomer and intradimer contacts and aremore rigid (Fig. 4a). The disorder of the N-terminiin solution was further supported by the analysis ofbackbone amide-exchange rates (data not shown).We suspect that at least some of these differencesare likely to result from the different solvent con-ditions used for crystallization and/or crystal-packing effects.

Nucleic-acid-binding sites of SARS-CoV NP CTD

Deletion studies revealed that residues 248–280are essential for the nucleic-acid-binding activityof the SARS-CoV NP CTD.5 However, the exactresidues involved in the binding had not been

identified. To identify these residues, we conductedchemical shift displacement (CSD) studies by titrat-ing 10-mer (dT10) or 20-mer (dT20) poly-deoxythy-mine (poly-dT) single-stranded DNA (ssDNA) intothe protein samples. ssDNA were used throughoutthis study as surrogates of single-stranded RNA.Titration of dT10 or dT20 into the protein samplecaused a concentration-dependent gradual shift ofsome resonances, instead of the appearance of a newset of resonances, suggesting that the binding occursin the fast-exchange regime, which is indicative of alow-affinity nucleic-acid-binding protein.13 FordT10, significant chemical shift changes were loca-lized primarily in the N-terminal region, particularlyK250, E253, A254, S256, K257, and K258 (Fig. 6a andb), while the majority of the other resonances werescarcely affected. This result suggests that dT10binds to the SARS-CoVNP at the N-terminal flexiblesegment, without affecting the overall structure ofthe protein (Fig. 4). The binding constant estimatedfrom the CSD studies at various dT10 concentrationsis Kd∼30 μM.Similarly, dT20 also generated significant chemical

shift changes in the same set of N-terminal residues;however, it also induced CSD of the resonances ofR320, H335, and A337, which are located in the β-sheet of the CTD dimer (Fig. 6b and d). Since theseresidues are not affected when the SARS-CoV NPCTD is bound to dT10 (Fig. 6a and c), we can rule outthe effect of long-range structural alterationsinduced by the binding of oligonucleotides to theN-termini. Our results suggest that R320, H335, andA337 also contribute to nucleic acid binding andcould be part of the binding site. This observation isunexpected, as these residues are sequentially andstructurally distant from the N-terminal region. Itshould be noted that we could only add dT20 upto a [dT20]/[CTD monomer] ratio of 1:4. A higherratio of DNA caused precipitation. As such, wecould not obtain a reliable dissociation constant forthe complex, and the chemical shift perturbationshown in Fig. 6b may not be the maximum changeexpected at a saturating dT20 concentration for thecomplex.

Mutagenesis of the nucleic-acid-binding sites ofSARS-CoV NP CTD

To further quantify the relative contribution ofpositively charged residues to oligonucleotide bind-ing by the SARS-CoV NP CTD, we produced doublemutants targeting K257/K258 and measured theeffect of the mutations on the apparent dissociationconstant (Kd) with fluorescently labeled dT20through EMSA.14 Since the mutation sites arelocated in the inherently flexible regions, the muta-tions did not cause any structural perturbations toother parts of the CTD dimer, as monitored by 15NHSQC spectra (Fig. 7). The EMSA results aresummarized in Table 2. We found that the charge-preserving K257R/K258R mutant did not affect theapparent binding affinity, compared to the wild-type construct, whereas the K257Q/K258Q mutant

Page 6: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Fig. 4. NMR structure of the SARS-CoV NP CTD. (a) Superposition of the 20 lowest-energy NMR structures of theSARS-CoV NP CTD and the corresponding crystal structure spanning residues 248–365.5 The two subunits in eachstructure are in orange and magenta for the NMR structure, and in red and green for the crystal structure. (b) Ribbondiagram of the solution structure of the SARS-CoV NP CTD. Secondary structure elements are labeled for one subunit.(c) Sequence of the SARS-CoV NP CTD. The secondary structures for the NMR structure and for the crystal structurespanning residues 248–365 are shown above the sequence, with red cylinders for α-helices and yellow arrows forβ-strands.

613Structure of SAIL SARS-CoV N Protein

showed a 5-fold reduction in affinity, althoughbinding was not completely abolished (Fig. 8, Table2). Our results suggest that the positive chargesat positions 257 and 258 of the SARS-CoV NP CTDare significant determinants of its binding affinitytowards oligonucleotides.Similarly, we also generated the R320A and

H335A mutants. The 15N HSQC spectra of thesemutants revealed that, in both cases, the structural

perturbations are mostly limited to the regionsadjacent to the Arg320 and His335 mutation sites(Fig. 9). However, the R320A mutation had achemical shift perturbation larger than that of theH335A mutation, probably because the Arg sidechain made more contacts with adjacent residues.The changes in the respective apparent dissociationconstants were measured by EMSA. Mutations atthis secondary binding site lowered the apparent

Page 7: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Fig. 5. Superposition of the NMR and crystal structures of a CTD monomer of SARS-CoV NP. The mean NMRstructure of a CTD monomer (blue) is superposed on the corresponding crystal structure spanning residues 248–365 (a)(red; PDB code 2cjr)5 and on that encompassing residues 270–370 (b) (light green; PDB code 2gib).7 In (a), the twostructures are superposed on the regions of residues 260–319 and 333–358, where the backbone RMSD between them is1.45 Å. In (b), the two structures are superposed on the regions of residues 274–319 and 333–358, where the backboneRMSD is 1.26 Å.

614 Structure of SAIL SARS-CoV N Protein

affinity towards dT20 by about twofold (Fig. 8, Table2). Although the effects were not as remarkableas those of the mutations near the N-terminus, theloss of binding affinity was still measurable. Ourresults are in agreement with the hypothesis that theβ-sheet region of the SARS-CoV NP CTD is part ofthe nucleic-acid-binding site.

Discussion

SAIL as an emergent tool for solution structuredetermination

The SAIL method is characterized by a sophisti-cated labeling pattern that is highly optimized forstructure determination, in contrast to other preex-isting isotope labeling techniques. Conventionalstrategies utilizing selective protonation under apredeuterated background lead to a compromisebetween increased intensity of the labeled protonsand loss of information about the deuterated ones.For instance, while the selective protonation of themethyl protons of Ile, Leu, and Val in a deuteratedbackground is very effective for the observation ofmethyl protons, one cannot obtain any informationon the remaining side chains.15 The SAIL method,on the other hand, combines the merits of increasedintensity through the deuteration of redundantprotons and preservation of the structural informa-tion by leaving key protons intact. Of particularinterest is the use of SAIL aromatic residues, whichwas first demonstrated with calmodulin.10 Assign-ments of chemical shift and NOE peaks involvingaromatic signals are indispensable for the high-quality structure determination of proteins, since

aromatic residues are often part of the folding core.In the case of the SARS-CoV NP CTD, the aromaticresonances of the UL protein were severely over-lapped, making their assignment difficult, if notimpossible (Fig. 3b and e). The introduction ofaromatic SAIL amino acids in the sample resolvedthis problem (Fig. 3d and f), ultimately leading to theelucidation of the SARS-CoV NP CTD solutionstructure. This study is the first case to havedemonstrated that the use of the SAIL phenylala-nine and tyrosine residues was effective in the NMRspectral analysis of a large protein. This is also thefirst instance of a homodimeric protein structure tohave been solved by the SAIL approach. The SAILmethod becomes more effective with increasingmolecular weight and allows for the structures oflarger proteins with even more intricate features tobe solved.8

Differences between the solution structure andthe crystal structures of SARS-CoV NP CTD

There are two distinct differences between thestructure of the SARS-CoV NP CTD in solution andthe structure of the SARS-CoV NP CTD in thecrystal. The first is the orientation of the short turn inthe β-sheet: in the crystal, the short turn is closer tothe N-terminal residues of the same protomer thanin the solution structure, resulting in a morecompact crystal structure (Fig. 5). This is observedin all of the crystal structures of the SARS-CoV NPCTD solved to date, regardless of the space groupand the construct.5,7 It is possible that crystalpacking is responsible for the compactness. On theother hand, the less compact solution structure ofthe short hairpin turn could be the result of the

Page 8: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Fig. 6. CSD of SARS-CoV NP CTD titrated with poly-dT ssDNA. Variation of the CSD of SARS-CoV NP CTD titrated with dT10 (a) or dT20 (b). The dashed lines in (a) and (b)represent the cutoff for significant displacements. (c) Spatial locations of residues (red) with CSD values larger than the cutoff value upon titration with dT10 (c) or dT20 (d). The twomonomers are in green and blue, respectively.

615Structure

ofSAIL

SARS-C

oVN

Protein

Page 9: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Fig. 7. Structure perturbation of K257/K258 double mutants. (a) Overlay of 15N-edited HSQC spectra from wild-typeCTD (blue), K257R/K258R (green), and K257Q/K258Q (red) double mutants. Affected resonances are identified by theirrespective residue types and numbers in the wild-type protein. These are mapped onto the ribbon structure of the CTDdimer in (b).

616 Structure of SAIL SARS-CoV N Protein

more dynamic character of the CTD in an aqueousenvironment.The second difference lies in the conformation of

the N-termini. The N-termini in the crystal structureof the SARS-CoV NP CTD, spanning residues 248–365, form an ordered conformation anchored by

intramolecular and intermolecular interactionsbetween various adjacent residues.5 However, theN-termini in solution are disordered and, in agree-ment with our previous studies,6 lack a short helixformed by residues 259–263 in the crystal structure(Fig. 4). The CTD is arranged as an octamer within

Page 10: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Table 2. Binding coefficients for dT20 to SARS-CoV NPCTD

Protein Apparent Kd (μM) Hill coefficient

Wild type 17.19±1.51 0.82±0.05K257R/K258R 13.96±0.92 0.92±0.05K257Q/K258Q 94.42±5.56 0.83±0.04R320A 33.81±2.16 0.85±0.04H335A 38.07±2.25 1.01±0.05

617Structure of SAIL SARS-CoV N Protein

the unit cell of the crystal, and this short helix couldbe the result of different solvent conditions, crystalpacking, and/or the oligomerization process.16

Residues within and adjacent to the short helix par-ticipate in intramolecular and intermolecular inter-actions within the crystal and contribute to theformation of the octamer.5 It is possible that theshort helix is selectively stabilized by the formationof new protein–protein contacts in the crystaloctamer, similar to those observed for the bindingof intrinsically disordered proteins to their targets.17Transient formation of the short helix is also ob-served in a few of the conformers within the NMRstructural ensemble.

Relevance to ribonucleoprotein packaging

In the crystal structure of SARS-CoVNP248–365, wehave previously found that the CTD forms an

Fig. 8. EMSA of SARS-CoV NP CTD mutants. (a) Mobil(K257/258R), K257Q/K258Q (K257/258Q), R320A, and H335Aby a factor of 2, starting from lane 1 (439 nM) to lane 11 (0.45K257/K258 double mutant towards dT20, compared to that ofH335A mutants towards dT20, compared to that of the wild-tyfrom three independent assays. Results are summarized in Ta

octamer.5 The packaging of the octamers in theasymmetric unit of the crystal results in two parallelbasic helical grooves. Residues 248–280 form apositively charged patch similar to that in theinfectious bronchitis virus NP.18 These patchesform a large part of the basic helical groove observedin the crystal structure. We postulate that the basichelical groove may serve as the RNA attachmentsite, and the structure suggests a mechanism forhelical RNA packaging in the virus. However, theoctamer has not been observed in solution. In ourDNA titration study, we found that the spin–spinrelaxation time, T2, of the amide resonancesdecreases, upon the addition of the DNA oligomer,at a rate faster than that expected for sheer increasesin molecular weight. We suspect that the DNAcomplex forms transient higher-order multimers insolution.Within the helical oligomer model, one expects

nucleic acid binding to stabilize the oligomer struc-ture. To investigate the consistency of our NMRchemical shift perturbation data with the proposedhelical model, we mapped the spatial locations ofthe residues perturbed by dT20 binding onto thehelical model proposed by Chen et al. (Fig. 10).5 Itclearly shows that both the N-terminal residues andthe additional perturbed residues in the β-sheetregion, namely, R320 and H335, form the interiorlining of the positively charged groove. The twoN-termini of the dimer reside on the outside edge

ity shift of dT20 bound to wild-type (wt), K257R/K258Rmutant proteins. The protein concentration was increasedmM). Lane C, negative control. (b) Binding curve of thethe wild-type protein. (c) Binding curve of the R320A andpe protein. Each curve in (b) and (c) represents the best fitble 2.

Page 11: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

‡www.Sail-technologies.com

618 Structure of SAIL SARS-CoV N Protein

and in the innermost part of the groove, respectively.The arrangement of the helical structure is such thatthe interior N-termini are still solvent-accessible.On the other hand, the perturbed β-sheet residuesreside in the midregion of the groove. Thus, whilethe current NMR titration results do not prove thevalidity of the helical model, they can still be satis-factorily accommodated within this framework.More rigorous data, such as the determination ofthe structure of the CTD/nucleic acid complex, arenecessary to provide definitive proof of the helicalmodel.

Oligonucleotide binding and structural disorder

We have previously shown that the SARS-CoVNPis a modular protein comprising two independentstructural domains connected by a 66-residue linkerand flanked on each end by the long, disordered N-and C-termini, which comprise 44 and 57 residues,respectively3 (Fig. 1a). The NTD, comprising resi-dues 45–181, has been shown to bind to RNA.4 Herewe showed that the CTD also binds to DNA withsimilar affinity to that of the NTD. We have shownpreviously that the di-domain fragment NP45-365binds to DNA and RNA with higher affinity thanthat of the respective NTD or CTD.15 Taken together,all of these observations suggest that the SARS-CoVNP binds to RNA at multiple sites, and the bindingstrength is enhanced by the multivalency effect, asmultiple binding sites are in contact with the RNAmolecule.19 This charge-based nonspecific bindingmode works in conjunction with intrinsic disorderto confer two main advantages to the nonspecificbinding of oligonucleotides to the CTD. First,because the disordered region is not locked into asingle conformation, binding to a variety of partnerscan occur regardless of the structural features ofthe partner, as long as the electrostatic interactionprovides enough free energy to maintain the boundstate. This property allows the CTD to bind to oligo-nucleotides with different sequences or tertiarystructures. This is an important feature of RNAchaperones, of which the SARS-CoV NP is amember, and hints at the possibility that residues248–270 are involved in the process.20,21 Second, theunstructured protein molecule can have a greatercapture radius for a specific binding site than that ofthe folded state with its restricted conformationalfreedom, the so-called “fly-casting mechanism.”22 Inthis binding scenario, the unfolded state bindsweakly at a relatively large distance, and then foldsas the protein approaches the binding site. These twoadvantages could act together to ensure that the CTDis able to bind to a variety of nucleotide sequenceswith enough affinity to carry out its function,namely, the encapsulation of the viral genome. Weenvision the extended conformation of the NPmolecule as a whole to facilitate its initial contactwith the RNA molecule in a fly-casting mechanism.Subsequent rearrangement of the NPmolecule in theRNA framework then results in favorable packing ofthe complex in a helical form.

Materials and Methods

Site-directed mutagenesis

The SARS-CoV NP CTD was cloned from SARS-CoVTW1 strain sequencing vectors (a gift from Dr. P.-J. Chen,National Taiwan University Hospital) as previouslydescribed.6 Mutants of the SARS-CoV NP CTD wereproduced with a QuickChange II kit (Stratagene, La Jolla,CA) on a RoboCycler 96 (Stratagene), in accordance withthe manufacturer's recommendations. Primers used formutagenesis were purchased from Mission Biotech(Taiwan). Mutations were confirmed through DNAsequencing.

Sample preparation

The SARS-CoV NP CTD, encompassing residues 248–365 including an extra MHHHHHHAMG sequence atthe N-terminus, was expressed in the E. coli BL21 (DE3)strain for nonlabeled and uniformly labeled samples, asdescribed previously,6 and in a cell-free reaction for theSAIL samples. The production of nonlabeled and uni-formly labeled samples by in vivo expression wasperformed in a conventional manner. The proteinsexpressed in E. coli were purified in accordance with ourpreviously described protocol.6 The cell-free expression ofthe SARS-CoV NP CTD was performed as describedpreviously.9 The S30 extract containing minimal residualamino acids was used for the cell-free expression. In thecell-free synthesis of the SARS-CoV NP CTD, the concen-tration of each SAIL amino acid was set to 0.5 mM, and2.3 mg of the SAIL-SARS-CoVNPCTDwas obtained froma total of 70 mg of SAIL amino mixture. SAIL amino acidswere obtained from SAIL Technologies, Inc.‡ The SAIL-SARS-CoV NP CTD thus produced was mainly in solubleform. The SAIL protein was purified by Ni-NTA affinitychromatography in 50mMsodiumphosphate (pH7.4) and150 mM NaCl, followed by gel filtration in a buffercontaining 50 mM sodium phosphate (pH 7.4), 150 mMNaCl, and 1 mM ethylenediaminetetraacetic acid (EDTA).The eluted SAIL-SARS-CoV NP CTD was then concen-trated and exchanged with the NMR buffer.

NMR spectroscopy

The SAIL-SARS-CoV NP CTD sample contained0.5 mM (10% D2O buffer) and 0.5 mM (100% D2O buffer)of the SAIL SARS-CoV NP CTD in NMR buffer [10 mMsodium phosphate pH 6.0, 50 mM NaCl, 1 mM EDTA,1 mM 2,2-dimethyl-2-silapentane-5-sulfonate, 0.01%NaN3, 10% D2O, and Complete Mini protease inhibitormix (Roche)]. SAIL-adapted NMR experiments for thestructure determination were performed at 30 °C withBruker 600-MHz or 800-MHz spectrometers equippedwith a TXI triple resonance room-temperature probe or acryoprobe. 1H–15N HSQC spectra were obtained with a 1-mM 15N-labeled sample in NMR buffer on a BrukerAvance 500-MHz spectrometer equipped with a TXIcryoprobe, using an in-house adaptation of the pulsesequence. For mutant characterization and protein–ssDNA-binding studies in NMR buffer, 15N-labeledsamples were prepared in NMR buffer, and spectra wererecorded on Bruker Avance 600-MHz or 800-MHz

Page 12: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Fig.

9(legend

onnext

page)

619Structure of SAIL SARS-CoV N Protein

Page 13: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

Fig. 10. Spatial locations of the nucleic-acid-binding sites in the helical packing model of the SARS-CoV NP CTDcrystal. (a) Binding sites are shown in CPK models, with the N-termini residues in magenta and with the residues on theβ-sheet (R320, H335, and A337) in green. The rest of the molecules are shown in a gray ribbon representation, except forthe β-sheets, which are in cyan. (b) Surface charge representation of the proposed helical supramolecular complex(adapted from Chen et al.5). The yellow and orange lines represent viral RNA strands. Notice that the binding sites in (a)are located in the positively charged grooves within the supramolecular complex.

620 Structure of SAIL SARS-CoV N Protein

spectrometers equipped with QXI quadruple resonance orTXI probes. The acquired data were processed with theXwinNMR suite (Bruker Biospin, Germany) or with iNMR(Nucleomatica, Italy), and chemical shift assignmentswere performed. The chemical shifts were referenced to2,2-dimethyl-2-silapentane-5-sulfonate and deposited inBioMagResBank.

Structure calculation and refinement

The NMR structure calculation of the SARS-CoV NPCTD was started using 28 independent pairs of inter-subunit distance restraints obtained from isotope-filteredNOE spectroscopy experiments reported previously.6

Automated NOE cross-peak assignments15 and structurecalculations with torsion-angle dynamics16 were per-formed using a modified version of the programCYANA 2.1, which incorporates SAIL labeling patterns,4

takes the homodimer symmetry explicitly into account forthe network anchoring of NOE assignments,15 ensures an

Fig. 9. Structure perturbation of R320A and H335A mutantype CTD (blue) and the R320A mutant (red). Affected resonnumbers in the wild-type protein. (b) Same as in (a), but with th(c) Mapping of residues affected by the R320A mutation (red)The side chains of R320 are shown in a neon representation. (dH335A mutation (in magenta). The side chain of H335 is also

identical conformation of the two monomers by imposingtorsion-angle difference restraints on all correspondingtorsion angles, and maintains a symmetric relativeorientation of the two monomers by applying distancedifference restraints between symmetry-related intermo-lecular Cα–Cα distances. Backbone torsion-angle restraintsobtained from database searches with the programTALOS17 were incorporated into the structural calcula-tion. Hydrogen-bond restraints were not used. CYANAstructure calculations were started from 100 randomizedconformers, and simulated annealing with 20,000 torsion-angle dynamics time steps per conformer was performed.The 20 conformers with the lowest final CYANA targetfunction values were subjected to restrained energy refine-ment in explicit solvent against the AMBER force field.18

CSD studies

A series of 2D 15N-edited HSQC spectra of uniformly15N-labeled SARS-CoV NP CTD protein (0.5 mM) was

ts. (a) Overlay of 15N-edited HSQC spectra from the wild-ances are identified by their respective residue types ande wild-type CTD (blue) and the H335Amutant (magenta).in the solution structure of the SARS-CoV NP CTD dimer.) Same as in (c), but showing the residues affected by theshown.

Page 14: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

621Structure of SAIL SARS-CoV N Protein

recorded in NMR buffer by titrating in different amountsof poly-dT ssDNA (Purigo, Taiwan). The affected amidecorrelations experienced CSD upon the addition ofssDNA. The unaffected and shifted resonances inuncrowded regions were easily assigned, whereas theshifted resonances in crowded regions were assigned bystepwise titration of the protein with small amounts ofssDNA and by tracing of the changes in the CSD until thedesired final concentration is achieved. The final protein/ssDNA ratio was 1:1 for 10-mer and 4:1 for 20-mer.Protein/ssDNA ratios higher than the one presented hereresulted in the formation of a precipitate within thesample. Theweighted CSD for each residuewas calculatedwith the formula: CSD=(1/2((Δδ1HN)

2+(Δδ15N/5)2)1/2,

where Δδ denotes the chemical shift difference betweenthe final complex and the free protein resonances. Theexperimental error in the weighted CSD from the spectralresolution was calculated as: (1/2((SW1

HN/points in1H)2+((SW15

N/points in15N)/5)2))1/2, where SW denotes

the total spectral width of the dimension. Amides withCSD values larger than the average shift of all the peaksplus the experimental error were selected as affected.

EMSA

All experiments were conducted in NMR buffer with 6-aminohexylfluorescein-labeled ssDNA (Purigo). Reac-tions were set up in 20-μl aliquots each containing50 nM 6-aminohexylfluorescein-labeled ssDNA. Proteinwas added to the aliquots starting at a concentration of500 μM, with each following aliquot containing a 2-foldserial dilution of the protein. A control reaction was setup where only ssDNA and buffer were added. Thealiquots were allowed to react at room temperature for30 min, and then were loaded on a 0.5× Tris–borateEDTA buffer DNA retardation gel (Invitrogen, Carlsbad,CA). The gel was run at 30 V and 4 °C for 2.5 h, and thebands were visualized with a Typhoon 9410 variablemode imager (Amersham Biosciences, Piscataway, NJ).Quantitation of the free ssDNA band was achievedthrough the ImageJ software (National Institutes ofHealth, Bethesda, MA). Bound ssDNA was estimatedby subtracting the free ssDNA band of each reaction fromthat of the control lane. The fraction of bound ssDNAwasfitted against the equation: Y=1/(1+(Kd/X)

n), usingGraphPad Prism (GraphPad Software, San Diego, CA),where Y is the fraction of ssDNA bound to the protein, Xis the protein concentration, Kd is the dissociationconstant, and n is the Hill coefficient. All experimentswere repeated twice.

PDB accession codes

Chemical shift assignments and atomic coordinateshave been deposited in BioMagResBank (accession code15511) and PDB (accession code 2jw8), respectively.

Acknowledgements

This work was supported, in part, by CoreResearch for Evolutional Science and Technology/Japan Science and Technology Agency (M.K. atTokyo Metropolitan University), by Technology

Development for Protein Analyses from Ministry ofEducation, Culture, Sports, Science and Technology(M.K. at Nagoya University), by a Grant-in-Aid forScientific Research of the Japan Society for thePromotion of Science (P.G. at Tokyo MetropolitanUniversity), and by theVolkswagen Foundation (P.G.at J.W. Goethe-University of Frankfurt am Main).This work was also supported by grants NSC 95-2311-B-001-066-MY3 (T.H.) and NSC-95-2113-M-001-035-MY3 (T.H.) from the National Science Council ofthe Republic of China. Some NMR spectra wereobtained at the High-Field Nuclear Magnetic Reso-nance Center, supported by the National ResearchProgram for Genomic Medicine, Taiwan, ROC.

References

1. Drosten, C., Gunther, S., Preiser, W., van der Werf, S.,Brodt, H. R., Becker, S. et al. (2003). Identification ofa novel coronavirus in patients with severe acuterespiratory syndrome. N. Engl. J. Med. 348, 1967–1976.

2. Ksiazek, T., Erdman, D., Goldsmith, C., Zaki, S., Peret,T., Emery, S. et al. (2003). A novel coronavirusassociated with severe acute respiratory syndrome.N. Engl. J. Med. 348, 1953–1966.

3. Chang, C., Sue, S., Yu, T., Hsieh, C., Tsai, C., Chiang, Y.et al. (2005). Modular organization of SARS corona-virus nucleocapsid protein. Biomed. Sci. 13, 59–72.

4. Huang, Q., Yu, L., Petros, A. M., Gunasekera, A., Liu,Z., Xu, N. et al. (2004). Structure of the N-terminalRNA-binding domain of the SARS CoV nucleocapsidprotein. Biochemistry, 43, 6059–6063.

5. Chen, C., Chang, C., Chang, Y., Sue, S., Bai, H., Riang,L. et al. (2007). Structure of the SARS coronavirusnucleocapsid protein RNA-binding dimerization do-main suggests a mechanism for helical packaging ofviral RNA. J. Mol. Biol. 368, 1075–1086.

6. Chang, C., Sue, S., Yu, T., Hsieh, C., Tsai, C., Chiang, Y.et al. (2005). The dimer interface of the SARS corona-virus nucleocapsid protein adapts a porcine respira-tory and reproductive syndrome virus-like structure.FEBS Lett. 579, 5663–5668.

7. Yu, I. M., Oldham, M. L., Zhang, J. & Chen, J. (2006).Crystal structure of the SARS coronavirus nucleo-capsid protein dimerization domain reveals evolu-tionary linkage between Corona- and Arteri-viridae.J. Biol. Chem. 281, 17134–17139.

8. Kainosho, M., Torizawa, T., Iwashita, Y., Terauchi, T.,Mei Ono, A. & Guntert, P. (2006). Optimal isotopelabelling for NMR protein structure determinations.Nature, 440, 52–57.

9. Torizawa, T., Shimizu, M., Taoka, M., Miyano, H. &Kainosho, M. (2004). Efficient production of isotopi-cally labeled proteins by cell-free synthesis: a practicalprotocol. J. Biomol. NMR, 30, 311–325.

10. Torizawa, T., Ono, A. M., Terauchi, T. & Kainosho, M.(2005). NMR assignment methods for the aromaticring resonances of phenylalanine and tyrosine resi-dues in proteins. J. Am. Chem. Soc. 127, 12620–12626.

11. Güntert, P., Mumenthaler, C. & Wüthrich, K. (1997).Torsion angle dynamics for NMR structure calculationwith the new program DYANA. J. Mol. Biol. 273,283–298.

12. Kabsch, W. & Sander, C. (1983). Dictionary of proteinsecondary structure: pattern recognition of hydrogen-bonded and geometrical features. Biopolymers, 22,2577–2637.

Page 15: 2008 Solution Structure of the C-terminal Dimerization Domain of SARS Coronavirus Nucleocapsid Protein Solved by the SAI

622 Structure of SAIL SARS-CoV N Protein

13. Dominguez, C. & Allain, F. (2006). NMR structureof the three quasi RNA recognition motifs (qRRMs)of human hnRNP F and interaction studies with Bcl-xG-tract RNA: a novel mode of RNA recognition.Nucleic Acids Res. 34, 3634–3645.

14. Garner, M. M. & Revzin, A. (1981). A gel electrophor-esis method for quantifying the binding of proteins tospecific DNA regions: application to components ofthe Escherichia coli lactose operon regulatory system.Nucleic Acids Res. 9, 3047–3060.

15. Tugarinov, V. & Kay, L. E. (2003). Ile, Leu, and Valmethyl assignments of the 723-residue malatesynthase G using a new labeling strategy and novelNMR methods. J. Am. Chem. Soc. 125, 13868–13878.

16. Andrec, M., Snyder, D. A., Zhou, Z., Young, J.,Montelione, G. T. & Levy, R. M. (2007). A largedata set comparison of protein structures deter-mined by crystallography and NMR: statisticaltest for structural differences and the effect ofcrystal packing. Proteins: Struct. Funct. Genet. 69,449–465.

17. Dyson, H. J. & Wright, P. E. (2005). Intrinsically un-structured proteins and their functions. Nat. Rev. Mol.Cell Biol. 6, 197–208.

18. Jayaram, H., Fan, H., Bowman, B. R., Ooi, A., Jayaram,J., Collisson, E. W. et al. (2006). X-ray structures of theN- and C-terminal domains of a coronavirus nucleo-capsid protein: implications for nucleocapsid forma-tion. J. Virol. 80, 6612–6620.

19. Lunde, B. M., Moore, C. & Varani, G. (2007). RNA-binding proteins: modular design for efficient func-tion. Nat. Rev., Mol. Cell Biol. 8, 479–490.

20. Zúñiga, S., Sola, I., Moreno, J., Sabella, P., Plana-Durán,J. & Enjuanes, L. (2007). Coronavirus nucleocapsidprotein is an RNA chaperone. Virology, 357, 215–227.

21. Tompa, P. & Csermely, P. (2004). The role of structuraldisorder in the function of RNA and protein chaper-ones. FASEB J. 18, 1169–1175.

22. Shoemaker, B. A., Portman, J. J. & Wolynes, P. G.(2000). Speeding molecular recognition by using thefolding funnel: the fly-casting mechanism. Proc. NatlAcad. Sci. USA, 97, 8868–8873.