6
Structural basis for suppression of a host antiviral response by influenza A virus Kalyan Das* , Li-Chung Ma* , Rong Xiao* , Brian Radvansky* , James Aramini* , Li Zhao* § , Jesper Marklund , Rei-Lin Kuo , Karen Y. Twu , Eddy Arnold* , Robert M. Krug , and Gaetano T. Montelione* ‡§ *Center for Advanced Biotechnology and Medicine and Departments of Chemistry and Chemical Biology and Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854; § Department of Biochemistry, Robert Wood Johnson Medical School, Piscataway, NJ 08854; and Institute for Cellular and Molecular Biology, Section of Molecular Genetics and Microbiology, University of Texas, Austin, TX 78712 Communicated by Aaron J. Shatkin, Center for Advanced Biotechnology and Medicine, Piscataway, NJ, June 11, 2008 (received for review March 24, 2008) Influenza A viruses are responsible for seasonal epidemics and high mortality pandemics. A major function of the viral NS1A protein, a virulence factor, is the inhibition of the production of IFN- mRNA and other antiviral mRNAs. The NS1A protein of the human influenza A/Udorn/72 (Ud) virus inhibits the production of these antiviral mRNAs by binding the cellular 30-kDa subunit of the cleavage and polyadenyl- ation specificity factor (CPSF30), which is required for the 3 end pro- cessing of all cellular pre-mRNAs. Here we report the 1.95-Å resolution X-ray crystal structure of the complex formed between the second and third zinc finger domain (F2F3) of CPSF30 and the C-terminal domain of the Ud NS1A protein. The complex is a tetramer, in which each of two F2F3 molecules wraps around two NS1A effector domains that interact with each other head-to-head. This structure identifies a CPSF30 binding pocket on NS1A comprised of amino acid residues that are highly conserved among human influenza A viruses. Single amino acid changes within this binding pocket eliminate CPSF30 binding, and a recombinant Ud virus expressing an NS1A protein with such a substitution is atten- uated and does not inhibit IFN- pre-mRNA processing. This binding pocket is a potential target for antiviral drug development. The crystal structure also reveals that two amino acids outside of this pocket, F103 and M106, which are highly conserved (>99%) among influenza A viruses isolated from humans, participate in key hydrophobic interac- tions with F2F3 that stabilize the complex. antiviral drug discovery bird flu vaccine engineering virology X-ray crystallography T he NS1 protein of human influenza A viruses (NS1A protein) is a small, multifunctional protein that participates in both protein-RNA and protein–protein interactions. Its N-terminal RNA-binding domain binds double-stranded RNA (dsRNA) (1–3). By identifying the replication defect of a recombinant influenza A/Udorn/72 (Ud) virus that encodes an NS1A protein lacking dsRNA-binding activity, it was established that the primary role of NS1A dsRNA-binding activity is the inhibition of the IFN-/- induced oligo A synthetase/RNase L pathway, and that NS1A dsRNA-binding activity has no detectable role in inhibiting the production of IFN- mRNA or inhibiting the activation of protein kinase R (PKR) (4, 5). The rest of the NS1A protein, which is referred to as the effector domain, has binding sites for several cellular proteins, including: the cellular 30-kDa subunit of the cleavage and polyadenylation specificity factor (CPSF30), a cellular factor required for the 3 end processing of cellular pre-mRNAs, thereby inhibiting the production of all cellular mRNAs, including IFN- mRNA (6–10); p85, resulting in the activation of phos- phatidylinositol-3-kinase signaling (11–14); and PKR, resulting in the inhibition of PKR activation (15). Of these multiple protein binding sites on the NS1A protein, only the dsRNA-binding site has been structurally characterized (3, 16–18). These structural studies have revealed key features of the NS1A dsRNA-binding site that can be targeted for the development of antivirals directed against influenza A virus (18). Here we describe the structure of the interface between CPSF30 and the NS1A protein, a molecular interaction that suppresses host antiviral responses. Specifi- cally, we report the 1.95-Å resolution X-ray crystal structure of the effector domain of the human influenza Ud NS1A protein in complex with a domain of CPSF30 comprising its second and third zinc (Zn) finger motifs (F2F3). We used the F2F3 domain of CPSF30 because it has been established that this domain binds efficiently to the Ud NS1A protein, and that expression of F2F3 in virus-infected cells leads to the inhibition of Ud virus replication and increased production of IFN- mRNA, presumably by occupying the CPSF30 binding site on the NS1A protein and hence blocking the binding of endogenous CPSF30 to this site (19, 20). This crystal structure reveals an NS1A:F2F3 tetrameric complex with two F2F3 binding pockets. The NS1A amino acids comprising the F2F3 binding pocket are highly conserved among human influenza A viruses, strongly suggesting that this CPSF30 binding pocket is used by all human influenza A viruses to suppress the production of IFN- mRNA. This binding pocket is a potential target for the development of antivirals directed against influenza A virus. The crystal structure also shows that the interaction surface between NS1A and F2F3 extends beyond the primary F2F3 binding pocket alone, and that two amino acids in the NS1A protein, Phe at position 103 and Met at position 106, play key roles in stabilizing the tetramer. Although F103 and M106 are highly conserved (99%) in the NS1A proteins of human influenza A viruses (21), a few prominent human inf luenza A viruses encode NS1A proteins with different amino acid residues at these positions. The biological properties of these few virus variants, however, reinforce the importance of NS1A protein-mediated CPSF30 binding for circu- lating human influenza A viruses. Results and Discussion The Crystal Structure Reveals a Tetrameric NS1A:F2F3 Complex. The Ud NS1A effector domain construct used in our experiments (amino acid residues 85-215) was identified by generation and assessment of the expression levels and solubility of 64 different NS1A constructs [supporting information (SI) Table S1]. This Ud NS1A (85-215) effector domain construct comprises 80% of the effector domain and is well-ordered in solution, as determined by its NMR spectra (Fig. S1). The 61-residue F2F3 tandem Zn-finger construct of CPSF30 comprises 30% of its full-length sequence, and is active in vivo in blocking interactions between full-length Ud Author contributions: K.D., L.-C.M., R.X., E.A., R.M.K., and G.T.M. designed research; K.D., L.-C.M., R.X., B.R., J.A., L.Z., J.M., R.-L.K., and K.Y.T. performed research; R.X., B.R., L.Z., J.M., R.-L.K., and K.Y.T. contributed new reagents/analytic tools; K.D., L.-C.M., R.X., B.R., J.A., J.M., R.-L.K., K.Y.T., E.A., R.M.K., and G.T.M. analyzed data; and K.D., L.-C.M., J.A., E.A., R.M.K., and G.T.M. wrote the paper. The authors declare no conflict of interest. Freely available online through the PNAS open access option. Data deposition: The atomic coordinates and structure factors have been deposited in the Protein Data Bank, www.pdb.org (PDB ID code 2RHK). To whom correspondence may be addressed. E-mail: [email protected], [email protected], or [email protected]. This article contains supporting information online at www.pnas.org/cgi/content/full/ 0805213105/DCSupplemental. © 2008 by The National Academy of Sciences of the USA www.pnas.orgcgidoi10.1073pnas.0805213105 PNAS September 2, 2008 vol. 105 no. 35 13093–13098 MICROBIOLOGY Downloaded by guest on February 3, 2022

Structural basis for suppression of a host antiviral

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Structural basis for suppression of a host antiviral

Structural basis for suppression of a host antiviralresponse by influenza A virusKalyan Das*†, Li-Chung Ma*‡, Rong Xiao*‡, Brian Radvansky*‡, James Aramini*‡, Li Zhao*§, Jesper Marklund¶,Rei-Lin Kuo¶, Karen Y. Twu¶, Eddy Arnold*†�, Robert M. Krug¶�, and Gaetano T. Montelione*‡§�

*Center for Advanced Biotechnology and Medicine and †Departments of Chemistry and Chemical Biology and ‡Molecular Biology and Biochemistry,Rutgers University, Piscataway, NJ 08854; §Department of Biochemistry, Robert Wood Johnson Medical School, Piscataway, NJ 08854; and¶Institute for Cellular and Molecular Biology, Section of Molecular Genetics and Microbiology, University of Texas, Austin, TX 78712

Communicated by Aaron J. Shatkin, Center for Advanced Biotechnology and Medicine, Piscataway, NJ, June 11, 2008 (received for review March 24, 2008)

Influenza A viruses are responsible for seasonal epidemics and highmortality pandemics. A major function of the viral NS1A protein, avirulence factor, is the inhibition of the production of IFN-� mRNA andother antiviral mRNAs. The NS1A protein of the human influenzaA/Udorn/72 (Ud) virus inhibits the production of these antiviral mRNAsby binding the cellular 30-kDa subunit of the cleavage and polyadenyl-ation specificity factor (CPSF30), which is required for the 3� end pro-cessing of all cellular pre-mRNAs. Here we report the 1.95-Å resolutionX-ray crystal structure of the complex formed between the second andthird zinc finger domain (F2F3) of CPSF30 and the C-terminal domain ofthe Ud NS1A protein. The complex is a tetramer, in which each of twoF2F3 molecules wraps around two NS1A effector domains that interactwith each other head-to-head. This structure identifies a CPSF30 bindingpocket on NS1A comprised of amino acid residues that are highlyconserved among human influenza A viruses. Single amino acid changeswithin this binding pocket eliminate CPSF30 binding, and a recombinantUd virus expressing an NS1A protein with such a substitution is atten-uated and does not inhibit IFN-� pre-mRNA processing. This bindingpocket is a potential target for antiviral drug development. The crystalstructure also reveals that two amino acids outside of this pocket, F103and M106, which are highly conserved (>99%) among influenza Aviruses isolated from humans, participate in key hydrophobic interac-tions with F2F3 that stabilize the complex.

antiviral drug discovery � bird flu � vaccine engineering � virology �X-ray crystallography

The NS1 protein of human influenza A viruses (NS1A protein)is a small, multifunctional protein that participates in both

protein-RNA and protein–protein interactions. Its N-terminalRNA-binding domain binds double-stranded RNA (dsRNA) (1–3).By identifying the replication defect of a recombinant influenzaA/Udorn/72 (Ud) virus that encodes an NS1A protein lackingdsRNA-binding activity, it was established that the primary role ofNS1A dsRNA-binding activity is the inhibition of the IFN-�/�-induced oligo A synthetase/RNase L pathway, and that NS1AdsRNA-binding activity has no detectable role in inhibiting theproduction of IFN-� mRNA or inhibiting the activation of proteinkinase R (PKR) (4, 5). The rest of the NS1A protein, which isreferred to as the effector domain, has binding sites for severalcellular proteins, including: the cellular 30-kDa subunit of thecleavage and polyadenylation specificity factor (CPSF30), a cellularfactor required for the 3� end processing of cellular pre-mRNAs,thereby inhibiting the production of all cellular mRNAs, includingIFN-� mRNA (6–10); p85�, resulting in the activation of phos-phatidylinositol-3-kinase signaling (11–14); and PKR, resulting inthe inhibition of PKR activation (15).

Of these multiple protein binding sites on the NS1A protein, only thedsRNA-binding site has been structurally characterized (3, 16–18).These structural studies have revealed key features of the NS1AdsRNA-binding site that can be targeted for the development ofantivirals directed against influenza A virus (18). Here we describe thestructure of the interface between CPSF30 and the NS1A protein, amolecular interaction that suppresses host antiviral responses. Specifi-

cally, we report the 1.95-Å resolution X-ray crystal structure of theeffector domain of the human influenza Ud NS1A protein in complexwith a domain of CPSF30 comprising its second and third zinc (Zn)finger motifs (F2F3). We used the F2F3 domain of CPSF30 because ithas been established that this domain binds efficiently to the Ud NS1Aprotein, and that expression of F2F3 in virus-infected cells leads to theinhibition of Ud virus replication and increased production of IFN-�mRNA, presumably by occupying the CPSF30 binding site on theNS1A protein and hence blocking the binding of endogenous CPSF30to this site (19, 20).

This crystal structure reveals an NS1A:F2F3 tetrameric complexwith two F2F3 binding pockets. The NS1A amino acids comprisingthe F2F3 binding pocket are highly conserved among humaninfluenza A viruses, strongly suggesting that this CPSF30 bindingpocket is used by all human influenza A viruses to suppress theproduction of IFN-� mRNA. This binding pocket is a potentialtarget for the development of antivirals directed against influenzaA virus. The crystal structure also shows that the interaction surfacebetween NS1A and F2F3 extends beyond the primary F2F3 bindingpocket alone, and that two amino acids in the NS1A protein, Pheat position 103 and Met at position 106, play key roles in stabilizingthe tetramer. Although F103 and M106 are highly conserved(�99%) in the NS1A proteins of human influenza A viruses (21),a few prominent human influenza A viruses encode NS1A proteinswith different amino acid residues at these positions. The biologicalproperties of these few virus variants, however, reinforce theimportance of NS1A protein-mediated CPSF30 binding for circu-lating human influenza A viruses.

Results and DiscussionThe Crystal Structure Reveals a Tetrameric NS1A:F2F3 Complex. TheUd NS1A effector domain construct used in our experiments(amino acid residues 85-215) was identified by generation andassessment of the expression levels and solubility of 64 differentNS1A constructs [supporting information (SI) Table S1]. This UdNS1A (85-215) effector domain construct comprises 80% of theeffector domain and is well-ordered in solution, as determined byits NMR spectra (Fig. S1). The 61-residue F2F3 tandem Zn-fingerconstruct of CPSF30 comprises �30% of its full-length sequence,and is active in vivo in blocking interactions between full-length Ud

Author contributions: K.D., L.-C.M., R.X., E.A., R.M.K., and G.T.M. designed research; K.D.,L.-C.M., R.X., B.R., J.A., L.Z., J.M., R.-L.K., and K.Y.T. performed research; R.X., B.R., L.Z., J.M.,R.-L.K., and K.Y.T. contributed new reagents/analytic tools; K.D., L.-C.M., R.X., B.R., J.A.,J.M., R.-L.K., K.Y.T., E.A., R.M.K., and G.T.M. analyzed data; and K.D., L.-C.M., J.A., E.A.,R.M.K., and G.T.M. wrote the paper.

The authors declare no conflict of interest.

Freely available online through the PNAS open access option.

Data deposition: The atomic coordinates and structure factors have been deposited in theProtein Data Bank, www.pdb.org (PDB ID code 2RHK).

�To whom correspondence may be addressed. E-mail: [email protected],[email protected], or [email protected].

This article contains supporting information online at www.pnas.org/cgi/content/full/0805213105/DCSupplemental.

© 2008 by The National Academy of Sciences of the USA

www.pnas.org�cgi�doi�10.1073�pnas.0805213105 PNAS � September 2, 2008 � vol. 105 � no. 35 � 13093–13098

MIC

ROBI

OLO

GY

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 3,

202

2

Page 2: Structural basis for suppression of a host antiviral

NS1A and full-length human CPSF30 (19). The complex betweenNS1A (85-215) and F2F3 was formed, purified by gel filtration (Fig.1A), and crystallized. The structure of this F2F3:NS1A (85-215)complex was then determined using selenomethionine (Se-Met)multiwavelength anomalous diffraction (MAD) techniques (22)and refined at 1.95-Å resolution, to Rwork and Rfree of 0.210 and0.234, respectively (Table S2). The chain fold of this Ud NS1Adomain is similar to that reported for the uncomplexed PR8 NS1Aeffector domain (PDB ID 2GX9) (23). Interestingly, the Zn fingersof F2F3 (Cys-X7/X8-Cys-X5/X4-Cys-X3-His) are structurally similarto the C3H Cys-X8-Cys-X5-Cys-X3-His Zn-finger domains of hu-man TIS11d, which binds class II AU-rich elements in the 3�untranslated regions of target mRNAs to regulate mRNA turnover(24). This structural similarity suggests a possible RNA-bindingfunction for these Zn-finger domains of CPSF30.

The structure of the F2F3:NS1A (85-215) complex reveals anunexpected mode of interaction between the NS1A protein andCPSF30. The complex is a tetramer, in which two F2F3 moleculeswrap around two NS1A effector domains that are interacting witheach other in a head-to-head orientation (Fig. 1B). The F2F3-binding surface has contributions from both chains of NS1A(85-215) in the head-to-head orientation (Fig. S2). The surface areaof one NS1A (85-215) molecule is �5,600 Å2, of which �1,680 Å2

participates in intermolecular interactions, while for each F2F3

molecule, �1,310 Å2 of �4,300 Å2 of surface area takes part intetramer formation.

The F2F3 domain of human CPSF30 used in our work (aminoacid residues 60-120) has a Ser at position 94, compared to thepublished sequence of CPSF30 (25), which has a Pro at this position.It is not clear if this single nucleotide variant (CCC to TCC) is anaturally occurring polymorphism or is a result of the cloningprocess. In any case, [S94]-F2F3 is biologically active in blockingCPSF30 binding by the NS1A protein in vivo, as it is the samemolecule that was used to demonstrate that F2F3 expression invirus-infected cells inhibits virus replication and increases virus-induced production of IFN-� mRNA (19, 20). Gel filtration datademonstrate that [S94]-F2F3 binds Ud NS1A (85-215), forming atetrameric complex with a molecular mass of �48 kDa (see Fig. 1A)similar to that obtained using [P94]-F2F3. Attempts to crystallizethe purified [P94]-F2F3:NS1A (85-215) complex provided only tinycrystals, while crystals of the [S94]-F2F3:NS1A (85-215) complex,with similar morphology, are significantly larger and suitable forX-ray crystallography. As illustrated in Fig. S3, the S94 residues inboth F2F3 molecules of the complex have proline-like backboneconformations (� � �72°; � � 172°), essentially identical to theconformation reported for residue P94 (� � �70°; � � 173°) in thesolution NMR structure of the isolated [P94]-F2F3 molecule (PDBaccession code 2D9N). In any case, the location of residue 94 in the

Fig. 1. Crystal structure of F2F3:NS1A (85-215) complex. (A) Gel filtration data demonstrating complex formation between NS1A (85-215) and F2F3. Traces showchromatographic profiles on a Superdex G75 column for NS1A (85-215) alone (red) and the complex of NS1A (85-215) with [S94]-F2F3 (blue). Inset showscalibrated gel filtration data for (A) [S94]-F2F3 (�10 kDa), (B) NS1A (85-215) alone (�27 kDa), and (C) [S94]-F2F3:NS1A complex (�47 kDa). Similar results wereobtained by static light scattering analysis of effluent fractions from size exclusion chromatography, as described in SI Materials and Methods: (A) 15 � 3 kDa,(B) 25 � 5 kDa, and (C) 48 � 5 kDa. The molecular mass expected for the tetrameric complex observed in the crystal structure is 49,250 Da, which is in goodagreement with these light scattering and gel filtration data. The elution times for isolated NS1A (85-215) (single chain calculated molecular mass 15,943 Da)and [S94]-F2F3 (single chain calculated molecular mass 8,682 Da) domains differ when loaded at different protein concentrations, suggesting that thesemolecules form weak homodimers under these solution conditions. Calibration standards (A–D) are described in SI Materials and Methods. (B) Two NS1A effectordomains (green and red) and two F2F3 domains (blue and yellow) of CPSF30 form the tetramer. Some NS1A� amino acid residues that function in complexformation are highlighted in cyan. (C) F3-binding pocket on NS1A (85-215). A hydrophobic pocket on the NS1A surface binds to the F3 Zn finger of F2F3. Bothchains of NS1A in the head-to-head dimer interact with each F2F3 molecule. (D) Expanded view of the F3-binding pocket. The NS1A amino acid residues labeledin red interact with the aromatic side chains of residues Y97, F98, and F102 of the F3 Zn finger of F2F3.

13094 � www.pnas.org�cgi�doi�10.1073�pnas.0805213105 Das et al.

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 3,

202

2

Page 3: Structural basis for suppression of a host antiviral

3D structure of the complex is distant from, and not in contact with,the NS1A effector domain (see Fig. S2). These results demonstratethat the S94 substitution does not disrupt the structure of F2F3, andthat [S94]-F2F3 and [P94]-F2F3 can form complexes with NS1A(85-215) with similar structures. Most importantly, as describedbelow, the [S94]-F2F3:NS1A (85-215) crystal structure accuratelypredicts effects of single-site mutations on specific functions of theNS1A protein in virus-infected cells, verifying the biological validityof this crystal structure.

The F2F3 Binding Pocket on the NS1A Protein. The crystal structureidentifies the F2F3 binding pocket on the surface of NS1A (Fig. 1C and D). This largely hydrophobic pocket, primarily defined byamino acid residues K110, I117, I119, Q121, V180, G183, G184, andW187, interacts with aromatic side chains of residues Y97, F98, andF102 of the F3 Zn finger of the corresponding F2F3 molecule.

To validate the biological relevance of the binding pocket,site-specific Ud NS1A protein variants were designed and evaluatedfor their effect on CPSF30-binding. Because the viral NS2/NEPprotein and NS1A are coded for by the same region of the viralgenome, but in a different translation frame (21), substitutions wereselected such that the amino acid sequence of NS2/NEP would notbe affected when a NS1A-mutant recombinant influenza A viruswas generated (described below); for example, only Arg substitu-tions are possible at the positions G184 or W187 of NS1A withoutaffecting the NS2/NEP sequence. Either of these two Arg substi-tutions, or substitution of Ala for Q121, eliminated detectablebinding of the full-length Ud NS1A protein to F2F3 in GST-pulldown experiments (Fig. 2A), confirming our hypotheses basedon the crystal structure that these three amino acids are requiredfor the formation of the F2F3:NS1A complex. To ascertain if theG184R substitution alters the structure of the NS1A protein,[G184R]-NS1A (85-215) was cloned, purified, and characterized.This amino acid substitution has little or no effect on the overall foldof NS1A (85-215), as indicated by amide circular dichroism and twodimensional (1H-15N)-HSQC NMR spectra (Fig. S4).

We next assessed the role of the CPSF30-binding pocket duringvirus infection by generating a recombinant Ud virus that expressesa NS1A protein with a G184R mutation. This recombinant virusforms plaques only �25% the size of WT plaques (Fig. 2B), andduring multiple cycle growth (at low multiplicity of infection) therecombinant replicates 20-fold slower than WT; for example, at 24 hafter infection the titers of recombinant and WT are 1.9 � 105 and3.8 � 106 pfu/ml, respectively. Attenuation of the G184R virus is notbecause of a reduction in the amount of the NS1A proteinsynthesized in G184R-infected cells (Fig. 2C). To determinewhether this attenuation is because of reduced suppression ofpre-mRNA processing, the relative amounts of IFN-� pre-mRNAand IFN-� mRNA in WT- and G184R-infected cells were deter-mined by quantitative RT-PCR (Fig. 2D). A substantial amount ofunprocessed IFN-� pre-mRNA was detected in WT virus-infectedcells, verifying that the Ud virus activates transcription of the IFN-�gene via the activation of interferon regulatory factor (IRF)-3 andother transcription factors (6, 9, 26). Approximately 20% as muchIFN-� pre-mRNA accumulated in G184R-infected cells, whereasthe amount of mature IFN-� mRNA was approximately five timesmore than that in WT-infected cells. Consequently, the processingof IFN-� pre-mRNA, which is largely blocked in WT virus-infectedcells, occurs much more efficiently in G184R virus-infected cells.This functional analysis demonstrates in vivo the biological signif-icance of the tetrameric [S94]-F2F3:NS1A (85-215) complex struc-ture, and particularly the importance of the CPSF30 binding pocket.It also provides definitive evidence for the essential role of NS1A-CPSF30 binding in the inhibition of IFN-� mRNA productionduring infection with influenza A/Udorn/72 virus.

Of the eight amino acid residues identified in the CPSF30-binding pocket of the Ud NS1A protein by this crystal structure, sixare almost completely (�98%) conserved among influenza Aviruses isolated from humans (21), strongly suggesting that thisCPSF30 binding site is used by all human influenza A viruses tosuppress the production of IFN-� mRNA. These residues are alsoconserved in H5N1 viruses isolated from humans and in thepandemic 1918 virus (A/Brevig Mission/1/18). The exceptions areI119 and V180, at the edge of the pocket shown in Fig. 1D, whichin some sequences are replaced by similar hydrophobic Met and Ileresidues, respectively, preserving the hydrophobicity of the pocket.

The Role of F103 and M106 of the NS1A Protein in Stabilizing theTetrameric Complex. The interaction surface between NS1A andF2F3 in the tetrameric complex extends beyond the primary F2F3binding pocket shown in Fig. 1D. Two NS1A amino acids outsidethe binding pocket, F103 and M106, are also critically involved information of the tetrameric complex (Fig. 3 A and B, and Fig. 4).As illustrated in Fig. 4, the side chain of residue M106 is positionedat the tetrameric epicenter and interacts with the side chain ofM106� of the NS1A� (molecule II) and with residues in both theF2F3 and F2F3� domains. The aromatic side chain F103 of NS1A(molecule I) interacts extensively with hydrophobic residues L72�,Y88�, and P111� of F2F3� (molecule II). Residues F103 and M106are required for the tight binding of F2F3 in vitro. Thus, for example,no observable binding between F2F3 and an Ud NS1A protein withsimultaneous F103L and M106I substitutions occurs in vitro GSTpull-down experiments (Fig. 3C).

In contrast, such F103L and M106I substitutions in the NS1Aprotein reduce, but do not completely eliminate, its binding toCPSF30 in infected cells because other viral proteins bind to andstabilize the NS1A:CPSF30 complex (20). This phenomenon wasobserved with the NS1A protein of the 1997 pathogenic H5N1influenza A/Hong Kong/483/97 (HK97) virus, which contains L(instead of F) at 103 and I (instead of M) at 106. The HK97 NS1Aprotein does not bind to F2F3 in vitro (20), like the mutant UdNS1A protein that contains L103 and I106 (see Fig. 3C), but doesbind CPSF30 to a significant extent in vivo when it is expressed ina virus that also encodes the other internal HK97 proteins (20). The

Fig. 2. Effects of amino acid substitutions in the NS1A protein on itsinteraction with CPSF30 and on its function in influenza A virus-infected cells.(A) GST-pulldown assay. GST-F2F3 or GST were mixed with equal amounts ofthe WT or indicated mutant 35S-labeled full-length NS1A protein of Ud, whichwere prepared as described in SI Materials and Methods. The labeled proteinseluted with glutathione from GST-F2F3 or GST were resolved by SDS-polyacrylamide gels, which were analyzed by exposure to X-ray film. (B)Plaque sizes of the WT and G184R mutant Ud viruses in Madin-Darby caninekidney (MDCK) cells. (C) The G184R mutation in the Ud NS1A protein does notaffect the amount of the NS1A protein synthesized in MDCK cells infected with5 pfu/cell. Immunoblots of cell extracts collected at 6 h after infection werecarried out with either anti-NS1A or antitubulin antibody. (D) QuantitativeRT-PCR measuring amounts of IFN-� pre-mRNA (Left) and IFN-� mRNA (Right)in WT and G184R Ud-infected cells. Pre-mRNA results were normalized to WT,and mRNA results were normalized to G184R data. The results show theaverage and standard deviation for the relative levels of G184R pre-mRNA andWT mRNA from three different virus infections.

Das et al. PNAS � September 2, 2008 � vol. 105 � no. 35 � 13095

MIC

ROBI

OLO

GY

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 3,

202

2

Page 4: Structural basis for suppression of a host antiviral

F103L and M106I mutations weaken, but do not prevent, complexformation in vivo because cognate HK97 internal proteins interactwith, and hence substantially stabilize, the CPSF30: HK97 NS1Acomplex in infected cells (20). Recent experiments show that such

binding of CPSF30 to the HK97 NS1A protein requires the HK97polymerase complex (PB1, PB2, PA, and NP), but not the HK97 Mprotein, and that the viral polymerase complex is actually part of theCPSF30:NS1A complex in infected cells, even when the NS1Aprotein has the optimum F103 and M106 amino acids (R.-L.K. andR.M.K., unpublished data).

Despite encoding a NS1A protein with less than optimumCPSF30 binding, the HK97 virus was pathogenic in birds, humans,mice, and ferrets (27, 28). Consequently, pathogenicity does notrequire a fully functional NS1A protein, and other viral proteins aresufficient to confer a pathogenic phenotype, consistent with a largebody of literature indicating that pathogenicity/virulence is poly-genic; that is, it cannot be ascribed to a single specific viral gene, butrather requires a combination of several, but not necessarily all, viralgenes (29–31). Nonetheless, attenuated CPSF30 binding by theHK97 virus is suboptimal for virulence: changing L103 to F andI106 to M results in not only a 20-fold enhancement in virusreplication in tissue culture (20), but also an even larger 250-fold,enhancement of virulence in mice (L.-M. Chen, R.T. Davis,R.-L.K., M. Malur, R.M.K., R.O. Donis, unpublished data). Thus,enhanced CPSF30 binding because of these two amino acid changesleads to enhanced influenza virulence in mice, demonstrating theimportance of the intermolecular interactions involving the highlyconserved F103 and M106 amino acids of the NS1A protein in thevirulence of influenza A viruses.

Unlike the NS1A protein of the HK97 virus, all of the NS1Aproteins of H5N1 viruses isolated from humans since 2003 containthe optimum F103 and M106 amino acids (20, 21). The origin of theselective pressure favoring replacement of F for L at 103 and of Mfor I at 106 in human H5N1 NS1A proteins is not known. Surpris-ingly, in 1999 to 2002, a period during which no H5N1 viruses wereisolated from humans, the vast majority of the H5N1 virusesisolated from avian species encoded NS1A proteins with F and Mat these positions (21). It is likely that these avian H5N1 viruseswere the source of the H5N1 viruses that were transmitted tohumans in 2003. It is not known what caused this change in the avianH5N1 NS1A protein, in light of the fact that the identities of the

Fig. 3. Structural role of F103 and M106 in formation of the tetramericcomplex. (A) Molecular graphics showing how two F2F3 molecules (repre-sented as the electrostatic potential surface) wrap around two NS1A (85-215)molecules. The head-to-head interaction of NS1A molecules forms a dockingsurface for F2F3 binding. The side chain of residue M106 is critically positionedat the tetrameric epicenter and interacts with the other three molecules. (B)Expanded regions showing the structural environments of amino acid residuesF103 and M106 of NS1A. The aromatic side chain of NS1A F103 interactsprimarily with the hydrophobic amino acid residues L72�, Y88� and P111� thatare present on the surface of F2F3�. (C) GST-pulldown assay. GST-F2F3 or GSTwas mixed with the WT or 103/106 mutant [F103L, M106I]-NS1A protein, andanalyzed as described in the legend of Fig. 2A.

Fig. 4. Molecular graphic showing the locations of NS1A residues F103 andM106 with respect to the F3-binding pocket. The tetrameric interface extendsbeyond the hydrophobic pocket of the NS1A effector domain (orange) whichbinds one F2F3 molecule (blue). Residues M106 and F103 (green surfaces) ofthe same NS1A effector domain interact with both the F2F3 (blue) and F2F3�(yellow) molecules. The M106 sidechain of this NS1A effector domain alsointeracts with the M106� side chain of the second NS1A� molecule (red) at thetetrameric epicenter.

13096 � www.pnas.org�cgi�doi�10.1073�pnas.0805213105 Das et al.

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 3,

202

2

Page 5: Structural basis for suppression of a host antiviral

amino acids at positions 103 and 106 in the NS1A proteins of othertypes of avian viruses (e.g., H9N2 and H6N1) showed considerablevariability from 1997 to 2006 (21).

F103 and M106 are highly conserved in seasonal (H1N1, H3N2,H2N2) human influenza A viruses. Of the 2,284 seasonal influenzaA viruses isolated from humans since 1933, 2,276 (99.6%) containF103 and M106 (21). Only four seasonal viruses have encoded aNS1A protein with Leu instead of the consensus Phe at position 103(21) and these viruses, like the H5N1 HK97 virus (20), presumablybind CPSF30 suboptimally in infected cells. Only five seasonalviruses have encoded an NS1A protein with a hydrophilic aminoacid (S) at position 103, namely, three viruses isolated in 1934 to1936 (including influenza A/PR/8/34), one virus isolated in 1954(A/Leningrad/54), and one virus isolated in 1976 (A/New Jersey/76)(21). The absence of such viruses since 1976 shows that influenzaA viruses encoding NS1A proteins with S103 are selected againstduring replication in humans.

Our F2F3:Ud NS1A structure predicts that a hydrophilic residueat position 103 in the NS1A protein should attenuate CPSF30binding (see Figs. 3B and 4), and indeed the A/PR/8/34 (PR8) NS1Aprotein that contains S103 does not bind CPSF30 in vitro (32). Inaddition, the PR8 NS1A protein does not inhibit cellular geneexpression in infected cells (32), indicating that it does not bindCPSF30 in infected cells (which we have confirmed). The 2.1 Åresolution crystal structure of the PR8 NS1A effector domain is adimer, stabilized by intermolecular �-sheet interactions (23). Asillustrated in Fig. S5, the oligomer orientations in the PR8 NS1Astructure are completely different from the F2F3-assisted head-to-head dimer of Ud NS1A effector domains observed in theF2F3:NS1A (85-215) complex structure. The Ud NS1A dimerformation is mediated by extensive hydrophobic interactions withtwo F2F3 molecules, whereas the PR8 NS1A dimer interfaceprimarily involves hydrogen bonds between two small �-strands(�1). The Ud NS1A effector domain also forms weak oligomers athigher concentrations and in the absence of F2F3 (as described inthe legend to Fig. 1). However, these interactions are weak andcould readily dissociate to form other oligomeric states with ap-propriate binding partners, such as the F2F3-stabilized head-to-head dimer seen in the Ud NS1A:F2F3 complex.

Why is CPSF30 binding by the PR8 NS1A protein not requiredduring virus infection? Previous studies have reported that PR8virus infection does not activate transcription factor IRF-3 (32, 33).In one set of experiments, IRF-3 activation was assayed by deter-mining whether the large amount of the NS1A protein that accu-mulated after 12 h of influenza A virus infection inhibited the IRF-3dimerization induced by a subsequent 6 h superinfection by Sendaivirus (32). Based on this assay, it was claimed that the NS1A proteinof PR8 virus, as well as the NS1A proteins of other influenza Aviruses, inhibits IRF-3 activation. However, it was not establishedthat the effect of such accumulated NS1A protein on subsequentSendai virus-induced IRF-3 activation accurately mirrors the ac-tions of the NS1A protein on IRF-3 activation induced by influenzaA virus itself at earlier times of infection. To address this issue, wedirectly assayed IRF-3 activation in influenza A virus-infected cellsby measuring the formation of the homodimer of IRF-3 that resultsfrom the phosphorylation-dependent activation of IRF-3 (6, 29).After infection of cells for 7 h with the Ud virus, �50% of theendogenous IRF-3 migrates in the position of the activated dimer(Fig. 5, lane 2), confirming our previous study (6). This activateddimeric IRF-3 functions to activate high level transcription of theIFN-� gene, as documented here by the accumulation of a sub-stantial amount of unprocessed IFN-� pre-mRNA in infected cells(see Fig. 2B). In fact, we have found that IRF-3 is activated byinfluenza A viruses expressing NS1A proteins encoded by manyother influenza A virus strains [e.g., two H5N1 viruses (HK97;A/Vietnam/1203/04) and the 1918 virus] (data not shown). Incontrast, the PR8 virus does not activate IRF-3 (see Fig. 5, lane 3),confirming previous studies by others (32, 33). Consequently,

because IRF-3 is not activated in PR8 virus-infected cells, and theresulting activation of IFN-� gene expression does not occur,CPSF30 binding by the PR8 NS1A protein is not required in PR8infected cells.

To determine whether this lack of IRF-3 activation is caused bythe PR8 NS1A protein itself, we generated a recombinant Ud virusthat expresses the PR8 instead of the Ud NS1A protein. In cellsinfected by this recombinant virus, IRF-3 is activated (Fig. 5, lane4), demonstrating that the PR8 NS1A protein does not suppressIRF-3 activation and that other mechanisms are responsible for thelack of IRF-3 activation in PR8 virus-infected cells. This recombi-nant virus is attenuated: during multiple cycle growth the recom-binant replicates 40-fold more slowly than WT; that is, at 24 h afterinfection the titers of the recombinant and Ud are 2 � 105 and 8 �106 pfu/ml, respectively. Despite encoding an NS1A protein that isdefective in CPSF30 binding (32), as well as defective in anotherfunction (34), the PR8 virus is pathogenic in mice (31), againattesting to the polygenic nature of pathogenicity (29–31).

ConclusionsThe X-ray crystal structure of the Ud NS1A (85-215):F2F3 complexdescribed here provides unique insights into the binding interfacebetween NS1A and CPSF30 and its linked suppression of a crucialhost antiviral response. These insights are not anticipated by theavailable structures of the F2F3 fragment alone (PDB ID 2D9N) orof the PR8 NS1A effector domain that does not bind F2F3 orCPSF30 (23, 32). The key structural features include two symmetricF2F3 binding pockets that are formed at a protein–protein interfacein the tetrameric structure of the Ud NS1A:F2F3 complex. The twoUd NS1A effector domains in this complex interact with each otherin a head-to-head orientation, which is unexpected and differentfrom the extended �-sheet dimer interface observed in the PR8NS1A effector domain (23). As illustrated in Fig. S6, this head-to-head dimer structure is also compatible with the known dimericstructure of the N-terminal RNA-binding domain (16, 17). The UdNS1A:F2F3 structure also explains the roles of NS1A residues F103and M106 in stabilizing the functional complex and their strongevolutionary conservation.

Based on these insights, we can conclude that CPSF30 binding bythe NS1A protein is the primary, if not the only, mechanism bywhich circulating human influenza A viruses suppress the produc-tion of IFN-� mRNA in infected cells. The X-ray crystal structurepresented here reveals the atomic details underlying this bindingprocess. Significantly, six of the amino acids comprising the F2F3/CPSF30-binding pocket are almost completely (�98%) conservedamong human influenza A viruses, including H5N1 viruses and the1918 virus (21). In addition, the two NS1A residues, F103 and

Fig. 5. PR8 virus infection, unlike infection by Ud virus, does not activateIRF-3. HEL299 cells were either mock-infected (M, lane 1), infected with 5pfu/cell of Ud virus (Ud, lane 2), infected with 5 pfu/cell of PR8 virus (PR8, lane3), or infected with 5 pfu/cell of a recombinant Ud virus in which the Ud NSgene was replaced by the PR8 NS gene (Ud/NS-PR8, lane 4). At 7 h afterinfection, cell extracts were prepared, subjected to electrophoresis on a 7.5%native gel, and IRF-3 monomers and dimers were detected by Western immu-noblotting using rabbit anti-IRF-3 antibody (36). An immunoblot with anti-NS1A antibody confirmed that equivalent amounts of the NS1A protein weresynthesized in Ud, PR8, and Ud/NS-PR8 virus-infected cells (lanes 2–4). Furtherdetails are provided in the SI Materials and Methods.

Das et al. PNAS � September 2, 2008 � vol. 105 � no. 35 � 13097

MIC

ROBI

OLO

GY

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 3,

202

2

Page 6: Structural basis for suppression of a host antiviral

M106, that stabilize the F2F3:NS1A tetrameric structure are alsoalmost completely conserved (�99%) among human influenza Aviruses (21). Interestingly, the properties of two prominent influ-enza A viruses, H5N1 HK97 and PR8, that encode NS1A proteinswith different amino acids at positions 103 and 106, reinforce theimportance of the CPSF30 binding site on the NS1A protein. TheHK97 NS1A protein contains different hydrophobic amino acids atpositions 103 and 106, and its binding to CPSF30 is stabilized ininfected cells via the interaction of the viral polymerase with theNS1A:CPSF30 complex (20), demonstrating that a supplementaryviral mechanism is used in infected cells to ensure that theNS1A:CPSF30 complex is formed. The PR8 NS1A protein has ahydrophilic (S) amino acid at position 103 that essentially eliminatesCPSF30 binding affinity, but PR8 virus uses a unique strategy tosuppress the production of IFN-� mRNA, namely suppressingIRF-3 activation by an undetermined mechanism. This PR8 strat-egy has been selected against during replication in humans, incompetition with influenza A viruses that both activate IRF-3 andrequire CPSF30 binding by the NS1A protein to suppress theproduction of mature IFN-� mRNA. This selection further dem-onstrates the crucial importance of NS1A protein-mediatedCPSF30 binding for circulating human influenza A viruses. Con-sidering that F2F3 expression in cells inhibits the replication ofinfluenza A viruses with no apparent effect on the cells (19), theintermolecular interfaces characterized here at atomic resolution,particularly the interface between the CPSF30 F3 finger andspecific conserved amino acid residues of NS1A (see Fig. 1), arecandidate target sites for the development of small-molecule anti-viral drugs.

Materials and MethodsNS1A effector domain NS1A (85-215) and the F2F3 (60-120) fragments of CPSF30,were cloned into modified pET21c and pET14c (Novagen) vectors (35). Theconstructs were verified by DNA sequence analysis and expressed in E. coliBL21(DE3) cells containing the rare tRNA expression plasmid pMGK. The over-expressed proteins were purified as described in SI Materials and Methods. Thecrystals of the purified [S94]-F2F3:NS1A complex were obtained by hanging dropvapor diffusion against the well solution containing 0.1 M sodium acetate pH 5.5,0.5 M KNO3, and 10% sucrose at 20°C. The structure was solved by Se-Met MADtechnique(22)andrefinedat1.95ÅresolutiontofinalRwork andRfree of0.210and0.234, respectively. Details of the X-ray crystallography methods are presented inthe SI Materials and Methods. Recombinant Ud viruses were generated fromcloned DNA as described in the SI Materials and Methods. IFN-� pre-mRNA andIFN-� mRNA in virus-infected cells were measured by real-time quantitativeRT-PCR as described in the SI Materials and Methods.

Note Added in Proof. While this paper was in press, another X-ray crystalstructure of an apo-NS1A effector domain was published (37), in which theNS1A dimer interface differs from that previously reported (as depicted in Fig.S5b). The dimer interface in this new structure involves residues which con-tribute to the F3-binding pocket in our structure (e.g., Trp-187).

ACKNOWLEDGMENTS. We thank T. Acton, A. Ertekin, Y.J. Huang, A. Shatkin,and C. Zhao for helpful discussions, and G. DeTitta from Hauptman-Woodward Medical Research Institute for the High-Throughput Crystalliza-tion Facility. This work was supported by institutional funds provided byRutgers University and by National Institutes of Health Grant AI11772 (toR.M.K.). Protein sample production was supported in part by the NortheastStructural Genomics Consortium of the National Institutes of Health ProteinStructure Initiative, Grant U54-074958 (to G.T.M.).

1. Hatada E, Takizawa T, Fukuda R (1992) Specific binding of influenza A virus NS1 proteinto the virus minus-sense RNA. in vitro J Gen Virol 73:17–25.

2. Wang W, et al. (1999) RNA binding by the novel helical domain of the influenza virusNS1 protein requires its dimer structure and a small number of specific basic aminoacids. RNA 5:195–205.

3. Chien CY, et al. (2004) Biophysical characterization of the complex between double-stranded RNA and the N-terminal domain of the NS1 protein from influenza A virus:evidence for a novel RNA-binding mode. Biochemistry 43:1950–1962.

4. Min J, Krug RM (2006) The primary function of RNA binding by the influenza A virusNS1 protein in infected cells: inhibiting the 2�-5� OAS/RNase L pathway. Proc Natl AcadSci USA 103:7100–7105.

5. Li S, Min JY, Krug RM, Sen GC (2006) Binding of the influenza A virus NS1 protein to PKRmediates the inhibition of its activation by either PACT or double-stranded RNA.Virology 349:13–21.

6. Kim MJ, Latham AG, Krug RM (2002) Human influenza viruses activate an interferon-independent transcription of cellular antiviral genes: outcome with influenza A virusis unique. Proc Natl Acad Sci USA 99:10096–10101.

7. Li Y, Chen ZY, Wang W, Baker CC, Krug RM (2001) The 3�-end-processing factor CPSFis required for the splicing of single-intron pre-mRNAs. in vivo RNA 7:920–931.

8. Nemeroff M, Barabino SML, Keller W, Krug RM (1998) Influenza virus NS1 proteininteracts with the 30 kD subunit of cleavage and specificity factor and inhibits 3� endformation of cellular pre-mRNAs. Mol Cell 1:991–1000.

9. Noah DL, Twu KY, Krug RM (2003) Cellular antiviral responses against influenza A virusare countered at the posttranscriptional level by the viral NS1A protein via its bindingto a cellular protein required for the 3� end processing of cellular pre-mRNAs. Virology307:386–395.

10. Shimizu K, Iguchi A, Gomyou R, Ono Y (1999) Influenza virus inhibits cleavage of theHSP70 pre-mRNAs at the polyadenylation site. Virology 254:213–219.

11. Hale BG, Jackson D, Chen YH, Lamb RA, Randall RE (2006) Influenza A virus NS1 proteinbinds p85beta and activates phosphatidylinositol-3-kinase signaling. Proc Natl AcadSci USA 103:14194–14199.

12. Shin YK, et al. (2007) SH3 binding motif 1 in influenza A virus NS1 protein is essentialfor PI3K/Akt signaling pathway activation. J Virol 81:12730–12739.

13. Shin YK, Liu Q, Tikoo SK, Babiuk LA, Zhou Y (2007) Influenza A virus NS1 proteinactivates the phosphatidylinositol 3-kinase (PI3K)/Akt pathway by direct interactionwith the p85 subunit of PI3K. J Gen Virol 88:13–18.

14. Hale BG, Batty IH, Downes CP, Randall RE (2008) Binding of influenza A virus NS1protein to the inter-SH2 domain of p85 suggests a novel mechanism for phosphoino-sitide 3-kinase activation. J Biol Chem 283:1372–1380.

15. Min JY, Li S, Sen GC, Krug RM (2007) A site on the influenza A virus NS1 protein mediatesboth inhibition of PKR activation and temporal regulation of viral RNA synthesis.Virology 363:236–243.

16. Chien CY, et al. (1997) A novel RNA-binding motif in influenza A virus non-structuralprotein 1 Nat Struct Biol 4:891–895.

17. Liu J, et al. (1997) Crystal structure of the unique RNA-binding domain of the influenzavirus NS1 protein. Nat Struct Biol 4:896–899.

18. Yin C, et al. (2007) Conserved surface features form the double-stranded RNA bindingsite of non-structural protein 1 (NS1) from influenza A and B viruses. J Biol Chem282:20584–20592.

19. Twu KY, Noah DL, Rao P, Kuo RL, Krug RM (2006) The CPSF30 binding site on the NS1Aprotein of influenza A virus is a potential antiviral target. J Virol 80:3957–3965.

20. Twu KY, Kuo RL, Marklund J, Krug RM (2007) The H5N1 influenza virus NS genesselected after 1998 enhance virus replication in mammalian cells. J Virol 81:8112–8121.

21. Macken C, Lu H, Goodman J, Boykin L (2001) in Options for the control of influenza IV,Osterhaus, NC, Hampson, AW, eds (Elsevier Science, Amsterdam), pp 103–106.

22. Hendrickson WA, Horton JR, LeMaster DM (1990) Selenomethionyl proteins producedfor analysis by multiwavelength anomalous diffraction (MAD): a vehicle for directdetermination of three-dimensional structure. EMBO J 9:1665–1672.

23. Bornholdt ZA, Prasad BV (2006) X-ray structure of influenza virus NS1 effector domain.Nat Struct Mol Biol 13:559–560.

24. Hudson BP, Martinez-Yamout MA, Dyson HJ, Wright PE (2004) Recognition of themRNA AU-rich element by the zinc finger domain of TIS11d. Nat Struct Mol Biol11:257–264.

25. Barabino SM, Hubner W, Jenny A, Minvielle-Sebastia L, Keller W (1997) The 30-kDsubunit of mammalian cleavage and polyadenylation specificity factor and its yeasthomolog are RNA-binding zinc finger proteins. Genes Dev 11:1703–1716.

26. Geiss GK, et al. (2002) Cellular transcriptional profiling in influenza A virus-infectedlung epithelial cells: the role of the nonstructural NS1 protein in the evasion of the hostinnate defense and its potential contribution to pandemic influenza. Proc Natl AcadSci USA 99:10736–10741.

27. Hatta M, Gao P, Halfmann P, Kawaoka Y (2001) Molecular basis for high virulence ofHong Kong H5N1 influenza A viruses. Science 293:1840–1842.

28. Zitzow LA, et al. (2002) Pathogenesis of avian influenza A (H5N1) viruses in ferrets.J Virol 76:4420–4429.

29. Horimoto T, Kawaoka Y (2005) Influenza: Lessons from past pandemics, warnings fromcurrent incidents. Nature Revs Microbiol 3:591–600.

30. Noah DL, Krug RM (2005) Influenza virus virulence and its molecular determinants. AdvVirus Res 65:121–145.

31. Wright PF, Webster RG (2001) in Fields Virology, Knipe, DM, Howley, PM, eds (Lippin-cott Williams & Wilkins, Philadelphia), pp 1533–1579.

32. Kochs G, Garcia-Sastre A, Martinez-Sobrido L (2007) Multiple anti-interferon actions ofthe influenza A virus NS1 protein. J Virol 81:7011–7021.

33. Talon J, et al. (2000) Influenza A and B viruses expressing altered NS1 proteins: Avaccine approach. Proc Natl Acad Sci USA 97:4309–4314.

34. Ozaki H, et al. (2004) Generation of high-yielding influenza A viruses in African greenmonkey kidney (Vero) cells by reverse genetics. J Virol 78:1851–1857.

35. Acton TB, et al. (2005) Robotic cloning and protein production platform of theNortheast Structural Genomics Consortium. Methods Enzymol 394:210–243.

36. Iwamura T, et al. (2001) Induction of IRF-3/-7 kinase and NF-kappaB in response todouble-stranded RNA and virus infection: common and unique pathways. Genes Cells6:375–388.

37. Hale BG, Barclay WS, Randall RE, Russell RJ (2008) Structure of an avian influenza A virusNS1 protein effector domain. Virology 378(1):1–5.

13098 � www.pnas.org�cgi�doi�10.1073�pnas.0805213105 Das et al.

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 3,

202

2