site-specific s-acylationofinfluenzavirushemagglutinin · hemagglutinin (ha) of influenza virus is...

13
Site-specific S-Acylation of Influenza Virus Hemagglutinin THE LOCATION OF THE ACYLATION SITE RELATIVE TO THE MEMBRANE BORDER IS THE DECISIVE FACTOR FOR ATTACHMENT OF STEARATE * Received for publication, June 3, 2014, and in revised form, October 12, 2014 Published, JBC Papers in Press, October 27, 2014, DOI 10.1074/jbc.M114.586180 Katharina Brett ‡1 , Larisa V. Kordyukova §1 , Marina V. Serebryakova §¶ , Ramil R. Mintaev § , Andrei V. Alexeevski § **, and Michael Veit ‡2 From the Institut für Virologie, Free University Berlin, 14163 Berlin, Germany, § A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia, I. I. Mechnikov Research Institute of Vaccines and Sera, Russian Academy of Medical Sciences, 105064 Moscow, Russia, and **Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia Background: S-Acylation of hemagglutinin with stearate and palmitate is essential for influenza virus replication. Results: Mass spectrometry showed that shifting a cysteine from the transmembrane region to a cytoplasmic position eliminates attachment of stearate. Conclusion: The location of the acylation site is the decisive factor for site-specific acylation. Significance: Similar differential acylation might occur in cellular transmembrane proteins. S-Acylation of hemagglutinin (HA), the main glycoprotein of influenza viruses, is an essential modification required for virus replication. Using mass spectrometry, we have previously dem- onstrated specific attachment of acyl chains to individual acyla- tion sites. Whereas the two cysteines in the cytoplasmic tail of HA contain only palmitate, stearate is exclusively attached to a cysteine positioned at the end of the transmembrane region (TMR). Here we analyzed recombinant viruses containing HA with exchange of conserved amino acids adjacent to acylation sites or with a TMR cysteine shifted to a cytoplasmic location to identify the molecular signal that determines preferential attachment of stearate. We first developed a new protocol for sample preparation that requires less material and might thus also be suitable to analyze cellular proteins. We observed cell type-specific differences in the fatty acid pattern of HA: more stearate was attached if human viruses were grown in mammalian compared with avian cells. No underacylated peptides were detected in the mass spectra, and even mutations that prevented generation of infectious virus particles did not abolish acylation of expressed HA as demonstrated by metabolic labeling experiments with [ 3 H]palmitate. Exchange of conserved amino acids in the vicinity of an acylation site had a moderate effect on the stearate content. In contrast, shifting the TMR cysteine to a cytoplasmic location virtually eliminated attachment of stearate. Thus, the location of an acylation site relative to the transmembrane span is the main signal for stearate attachment, but the sequence context and the cell type modulate the fatty acid pattern. Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep- tide, a large ectodomain, a single transmembrane region (TMR 3 ; 27 amino acids) and an 11-amino acid-long cytoplas- mic tail (1). HA from influenza A virus is S-acylated at three cysteine residues, two located in the cytoplasmic domain and one located at the end of the TMR (2– 4). The hydrophobic modification of HA is essential for virus replication because (depending on the virus strain) virus mutants with more than one acylation site deleted either show drastically impaired growth or could not be created at all by reverse genetics (5–7). Acylation facilitates raft association of HA (8 –10) and thus enrichment of the protein in small nanodomains of the plasma membrane (11), an observation that might explain why palmi- toylation affects both assembly of virus particles (5, 7, 12) and the membrane fusion activity of HA (2, 6, 13–15). In accord- ance with the essential requirement of palmitoylation of HA for virus replication is the complete conservation of the acylation sites through all HA subtypes and variants, although HA is a highly variable molecule with very low amino acid conservation (16). Cellular enzymes that acylate HA (or any other viral protein) have not been identified, but likely candidates are members of the Asp-His-His-Cys (DHHC) family, polytopic membrane proteins containing a DHHC motif within one of their cytoplas- mic domains (17, 18). Most of the DHHC proteins are located in the endoplasmic reticulum or Golgi region where acylation of HA takes place (19). Alternatively, S-acylation of at least some proteins can occur by a non-enzymatic or autocatalytic mech- anism (20, 21). * This work was supported by Deutsche Forschungsgemeinschaft Grant Ve 141/10 and Priority Program 1175 and Russian Foundation for Basic Research Grants 13-04-00290 and 14-04-01693. 1 Both authors contributed equally to this work. 2 To whom correspondence should be addressed: Inst. für Virologie, Fachbe- reich Veterinärmedizin, Zentrum für Infektionsmedizin-Robert-von-Os- tertag-Haus, Freie Universität Berlin, Robert-von-Ostertag-Strasse 7-13, 14163 Berlin, Germany. Tel.: 49-30-838-51891; Fax: 49-30-838-451847; E-mail: [email protected]. 3 The abbreviations used are: TMR, transmembrane region; DHHC protein, polytopic membrane protein containing an Asp-His-His-Cys motif; MDCK, Madin-Darby canine kidney; TPCK, L-1-tosylamido-2-phenylethyl chloro- methyl ketone; NP, nucleoprotein; Pal, palmitate; Stear, stearate; CEF, chicken embryo fibroblast. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 50, pp. 34978 –34989, December 12, 2014 © 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. 34978 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 50 • DECEMBER 12, 2014 by guest on December 4, 2020 http://www.jbc.org/ Downloaded from

Upload: others

Post on 21-Aug-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

Site-specific S-Acylation of Influenza Virus HemagglutininTHE LOCATION OF THE ACYLATION SITE RELATIVE TO THE MEMBRANE BORDER IS THEDECISIVE FACTOR FOR ATTACHMENT OF STEARATE*

Received for publication, June 3, 2014, and in revised form, October 12, 2014 Published, JBC Papers in Press, October 27, 2014, DOI 10.1074/jbc.M114.586180

Katharina Brett‡1, Larisa V. Kordyukova§1, Marina V. Serebryakova§¶, Ramil R. Mintaev§�, Andrei V. Alexeevski§**,and Michael Veit‡2

From the ‡Institut für Virologie, Free University Berlin, 14163 Berlin, Germany, §A. N. Belozersky Institute of Physico-ChemicalBiology, Lomonosov Moscow State University, 119991 Moscow, Russia, ¶Institute of Gene Biology, Russian Academy of Sciences,119334 Moscow, Russia, �I. I. Mechnikov Research Institute of Vaccines and Sera, Russian Academy of Medical Sciences,105064 Moscow, Russia, and **Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University,119234 Moscow, Russia

Background: S-Acylation of hemagglutinin with stearate and palmitate is essential for influenza virus replication.Results: Mass spectrometry showed that shifting a cysteine from the transmembrane region to a cytoplasmic position eliminatesattachment of stearate.Conclusion: The location of the acylation site is the decisive factor for site-specific acylation.Significance: Similar differential acylation might occur in cellular transmembrane proteins.

S-Acylation of hemagglutinin (HA), the main glycoprotein ofinfluenza viruses, is an essential modification required for virusreplication. Using mass spectrometry, we have previously dem-onstrated specific attachment of acyl chains to individual acyla-tion sites. Whereas the two cysteines in the cytoplasmic tail ofHA contain only palmitate, stearate is exclusively attached to acysteine positioned at the end of the transmembrane region(TMR). Here we analyzed recombinant viruses containing HAwith exchange of conserved amino acids adjacent to acylationsites or with a TMR cysteine shifted to a cytoplasmic location toidentify the molecular signal that determines preferentialattachment of stearate. We first developed a new protocol forsample preparation that requires less material and might thusalso be suitable to analyze cellular proteins. We observed celltype-specific differences in the fatty acid pattern of HA: morestearate was attached if human viruses were grown in mammaliancompared with avian cells. No underacylated peptides weredetected in the mass spectra, and even mutations that preventedgeneration of infectious virus particles did not abolish acylation ofexpressed HA as demonstrated by metabolic labeling experimentswith [3H]palmitate. Exchange of conserved amino acids in thevicinity of an acylation site had a moderate effect on the stearatecontent. In contrast, shifting the TMR cysteine to a cytoplasmiclocation virtually eliminated attachment of stearate. Thus, thelocation of an acylation site relative to the transmembrane span isthe main signal for stearate attachment, but the sequence contextand the cell type modulate the fatty acid pattern.

Hemagglutinin (HA) of influenza virus is a typical type Itransmembrane glycoprotein with an N-terminal signal pep-tide, a large ectodomain, a single transmembrane region(TMR3; 27 amino acids) and an 11-amino acid-long cytoplas-mic tail (1). HA from influenza A virus is S-acylated at threecysteine residues, two located in the cytoplasmic domain andone located at the end of the TMR (2– 4). The hydrophobicmodification of HA is essential for virus replication because(depending on the virus strain) virus mutants with more thanone acylation site deleted either show drastically impairedgrowth or could not be created at all by reverse genetics (5–7).Acylation facilitates raft association of HA (8 –10) and thusenrichment of the protein in small nanodomains of the plasmamembrane (11), an observation that might explain why palmi-toylation affects both assembly of virus particles (5, 7, 12) andthe membrane fusion activity of HA (2, 6, 13–15). In accord-ance with the essential requirement of palmitoylation of HA forvirus replication is the complete conservation of the acylationsites through all HA subtypes and variants, although HA is ahighly variable molecule with very low amino acid conservation(16).

Cellular enzymes that acylate HA (or any other viral protein)have not been identified, but likely candidates are members ofthe Asp-His-His-Cys (DHHC) family, polytopic membraneproteins containing a DHHC motif within one of their cytoplas-mic domains (17, 18). Most of the DHHC proteins are located inthe endoplasmic reticulum or Golgi region where acylation ofHA takes place (19). Alternatively, S-acylation of at least someproteins can occur by a non-enzymatic or autocatalytic mech-anism (20, 21).* This work was supported by Deutsche Forschungsgemeinschaft Grant Ve

141/10 and Priority Program 1175 and Russian Foundation for BasicResearch Grants 13-04-00290 and 14-04-01693.

1 Both authors contributed equally to this work.2 To whom correspondence should be addressed: Inst. für Virologie, Fachbe-

reich Veterinärmedizin, Zentrum für Infektionsmedizin-Robert-von-Os-tertag-Haus, Freie Universität Berlin, Robert-von-Ostertag-Strasse 7-13,14163 Berlin, Germany. Tel.: 49-30-838-51891; Fax: 49-30-838-451847;E-mail: [email protected].

3 The abbreviations used are: TMR, transmembrane region; DHHC protein,polytopic membrane protein containing an Asp-His-His-Cys motif; MDCK,Madin-Darby canine kidney; TPCK, L-1-tosylamido-2-phenylethyl chloro-methyl ketone; NP, nucleoprotein; Pal, palmitate; Stear, stearate; CEF,chicken embryo fibroblast.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 50, pp. 34978 –34989, December 12, 2014© 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

34978 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 50 • DECEMBER 12, 2014

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 2: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

It has long been known that some “palmitoylated” proteinscontain different acyl chains, indicating that the responsibleenzyme(s) (in contrast to the N-myristoyltransferase) cannotstrictly discriminate between long chain fatty acid species (22).However, advancements in mass spectrometry have allowedprecise quantification of the type of fatty acid linked to an acylprotein or even to a single acylation site. The peripheral mem-brane protein GAP43 contains both palmitate (C16:0) and ste-arate (C18:0) at two N-terminal cysteine residues, but a prefer-ence of a fatty acid species for one of the cysteines was notobserved (23). In contrast, our studies with HAs of influenza Avirus revealed that stearate is exclusively attached to the cys-teine positioned at the end of the transmembrane region,whereas the two cytoplasmic cysteines contain only palmitate(24). Site-specific acylation was also observed for the glycopro-teins of influenza B and C viruses. HA of influenza B virus pos-sesses two cytoplasmic cysteines that contain only palmitate,whereas the hemagglutinin esterase fusion protein HEF ofinfluenza C virus has one transmembrane cysteine that isstearoylated (24), confirming previous studies performed withless sophisticated methodology (25). We also analyzed glyco-proteins of other enveloped viruses and found that site-specificattachment of palmitate or stearate is a common feature of viralspike proteins (26) and (because viruses rely on the cellularacylation machinery) probably also of cellular transmembranereceptors.

Since this initial observation, we have analyzed more than 40HA variants from 14 subtypes by MS (27, 28). The percentage ofstearate in all HAs differs from 35% (suggesting that each of theTMR cysteines of the trimeric HA spike contains stearate) to12% (indicating that only one of three TMR cysteines is stearoy-lated). Interestingly, HA present in virus strains initially iso-lated from humans contain less stearate compared with HAisolated from mammals and especially from birds. Because allour previous MS analyses were performed with viruses purifiedfrom the allantoic fluid of embryonated chicken eggs, it is notknown whether cell type-specific differences in the fatty acidpattern might exist. Another reason for the different stearatecontent in the HA of human and avian viruses might be thatother viral membrane proteins, especially the matrix proteinM1, which is much less variable than HA but contains host-specific amino acid substitutions, affect acylation of HA. How-ever, we could not detect variability in the stearate content ofHA if internal proteins were exchanged between viruses (29).

Thus, the main molecular signal that determines preferentialattachment of palmitate or stearate is likely to be located in HAeither in the amino acid sequence around individual acylationsites or in their positioning relative to the membrane span. Toanalyze this question, we created recombinant viruses contain-ing HA with point mutations adjacent to acylation sites or witha TMR cysteine shifted to a cytoplasmic location and analyzedthe fatty acid content of HA by MS utilizing a new procedure forsample preparation that is more suitable for viruses grown incell culture instead of embryonated eggs.

EXPERIMENTAL PROCEDURES

Cells and Plasmids—African green monkey kidney cells(CV1), human embryonic kidney cells (293T), DF-1 cells, a

chicken fibroblast cell line, and Madin-Darby canine kidney(MDCK) II cells were maintained in Dulbecco’s modifiedEagle’s medium (DMEM; Pan Biotech) supplemented with 10%fetal calf serum, 100 units/ml penicillin, and 100 �g/ml strep-tomycin. Chicken embryo fibroblasts were prepared fromchicken embryos as described (30). All cells were cultured at37 °C in 5% CO2 and passaged twice per week. The plasmidpHW2000 used for the rescue of recombinant influenzaA/WSN/33 (H1N1) virus has been described (31).

Generation of Recombinant Virus—Recombinant influenzaA/WSN/33 (H1N1) virus (WSN) was generated using theeight-plasmid reverse genetics system in which each plasmidcontains cDNA of one viral RNA segment flanked by suitablepromoters. In the HA-encoding cDNA segment, the codon forIle-563 (ATA) was replaced by the isoleucine codon TTG, andGly-557 (GGG) was replaced by the alanine codon GCT and theglutamic acid codon GAA, respectively. Gln-560 (CAG) wasreplaced by the glutamine codon GAA. Gly-547 (GGG) wasreplaced by the serine codon ACG. Cys-554 (TGT) and Leu-559(TTG) were replaced by the serine codon (AGC) and the cys-teine codon (TGC), respectively, using QuikChange mutagen-esis (Stratagene) and confirmed by sequencing (GATC Bio-tech). 293T cells in 60-mm dishes were transfected with eightplasmids encoding WSN cDNA (1 �g each) using TurboFect(Fermentas/Thermo) in Opti-MEM (Invitrogen/Thermo).4 – 6 h after transfection, medium was replaced by infectionmedium (DMEM containing 0.2% bovine serum albumin, 0.1%FBS, 2 mM glutamine, 100 units/ml penicillin/streptomycin,and 1 �g/ml TPCK-treated trypsin (Sigma-Aldrich), and incu-bation was continued at 37 °C. 72 h after transfection, superna-tants were harvested and cleared of debris by low speed centri-fugation (2,000 � g, 5 min, 4 °C). Recombinant viruses were firstplaque-purified on MDCKII cells and then amplified andtitrated in MDCKII cells to obtain a high titer virus stock.

Plaque Assay—The plaque assay was performed on MDCKIIcells in 6-well plates according to standard procedures. Briefly,cells were infected with serial 10-fold dilutions of the virussupernatants in infection medium, incubated for 1 h at 37 °C,washed twice with phosphate-buffered saline (PBS), and over-laid with 1.8% SeaPlaque agarose (Lonza) in 2� Eagle’s mini-mum essential medium (Lonza) supplemented with 0.2%bovine serum albumin, 0.1% FBS, 2 mM glutamine, and 1 �g/mlTPCK-treated trypsin. After 2–3 days of incubation, cells werestained using neutral red (0.03% in PBS; Biochrom), andplaques were counted.

For isolation of virus clones, the overlaying agar of single,well separated plaques was excised, dissolved in 500 �l of PBS,incubated for 16 h at 4 °C, and then used to infect T25 cellculture flasks of MDCKII cells. After incubation for 1 h at 37 °C,the cells were washed twice with PBS, and infection mediumwas added. At 1–3 days postinfection when a cytopathic effectwas visible, supernatants were harvested and cleared of debrisby low speed centrifugation (2,000 � g, 5 min, 4 °C). Every virussample was sequenced to confirm the presence of insertedmutations before titrating in the plaque assay and amplificationin large scale in MDCKII cells.

Differential Acylation of Influenza HA

DECEMBER 12, 2014 • VOLUME 289 • NUMBER 50 JOURNAL OF BIOLOGICAL CHEMISTRY 34979

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 3: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

Virus Preparation from Embryonated Eggs and MDCKCells—Recombinant WSN virus (1,000 pfu; diluted in 0.9%NaCl) was inoculated with a 0.45 � 25-mm needle into theallantoic cavity of 11-day-old embryonated chicken eggs. Forcontrol experiments, natural virus strains A/WSN/33 (H1N1)and A/Puerto Rico/8/34 (H1N1) (PR8) were propagated in the11-day-old embryonated chicken eggs via the same procedure.After 48 –72 h at 37 °C, the embryos were euthanized at 4 °C for16 h. The allantoic fluid was collected and centrifuged at2,000 � g for 20 min at 4 °C to clarify from debris anderythrocytes.

To prepare virus from cell culture, the virus stock was used toinfect MDCKII, chicken embryo fibroblast (CEF), or DF-1 cellswith a multiplicity of infection of 0.01. After 1 h of adsorption,cells were washed with PBS, infection medium was supplied,incubation was continued until a cytopathic effect became evi-dent, and then the supernatant was harvested.

The cleared supernatant or allantoic fluid was centrifuged at100,000 � g for 2 h, and the viral pellet was resuspended in TNE(10 mM Tris/HCl, pH 7.4, 100 mM NaCl, and 1 mM EDTA). Insome cases, the obtained pellet was centrifuged through 2 ml ofa sucrose cushion (20% in TNE) solution and centrifuged at100,000 � g for 2 h. The resulting virus pellet was resolved inTNE.

Reverse Transcription-Polymerase Chain Reaction (RT)-PCRand Sequencing—To check for correctness of HA sequences inthe recombinant viruses, RNA was isolated from cell culturesupernatants or virus preparations with an Invisorb Spin VirusRNA Mini kit (Stratec) followed by RT-PCR using HA-specificprimers and a OneStep RT-PCR kit (Qiagen). PCR productswere purified from agarose gels using the Fragment CleanUp kit(Stratec) and sequenced (GATC Biotech).

Expression of HA with the Vaccinia T7 System and MetabolicLabeling—HA was subcloned from pHW2000 constructs intothe plasmid pTM1 behind the T7 RNA polymerase promoter.CV1 cells (2 � 106) in Petri dishes were infected with recombi-nant vaccinia virus expressing T7 polymerase (10 plaque-form-ing units/cell). At 2 h postinfection, the cells were washed withPBS prior to transfection with pTM1-HA plasmids usingTurboFect. At 5 h postinfection, cells intended for [35S]methio-nine/cysteine labeling were starved in DMEM lacking bothamino acids. Cells were labeled at 5.5 h postinfection with 0.5mCi/ml [35S]methionine/cysteine (1,175 Ci/mmol; Easy TagExpress Protein Labeling Mix, PerkinElmer Life Sciences) orwith 0.25 mCi/ml [3H]palmitic acid (30 – 60 Ci/mmol; 9,10-3H;PerkinElmer Life Sciences) for 3 h. Cells were lysed in radioim-mune precipitation assay buffer (20 mM Tris-HCl, 150 mM

NaCl, 10 mM EDTA, 10 mM iodoacetamide, 0.1% (w/v) SDS, 1%(v/v) Triton X-100, and 1% (w/v) sodium deoxycholate, pH 7.4),cell debris was pelleted, and the cleared supernatant was incu-bated at 4 °C for 16 h with 1 �l of anti-influenza H1N1 viruspolyclonal rabbit antibody R309 (32) kindly provided by Y.Kawaoka. Following incubation with 30 �l of protein A-Sephar-ose, each lysate was washed with radioimmune precipitationassay buffer. The final pellet was analyzed by SDS-PAGE undernon-reducing conditions and fluorography as described (33).Densitometric quantification of bands was carried out usingBio-1D software (Vilmer-Lourmat).

Mass Spectrometry: Sample Preparation by Proteolytic Diges-tion of Virus Particles—has been described (34). Briefly, the HAectodomain was removed by digestion of gradient-purified virusparticles with bromelain, shaved particles were pelleted, and themembrane-anchoring fragments of HA were extracted to thechloroform phase and analyzed directly by MALDI-TOF MS.

Sample Preparation by Blotting—Viral proteins were sepa-rated via non-reducing SDS-PAGE in a Mini-Protean (Bio-Rad)system and electroblotted onto a PVDF membrane (Merck),which was stained with Ponceau S (Fluka). The HA band was cutfrom the membrane, fragmented into small pieces, and stored at4 °C in an Eppendorf tube with � 50 �l of distilled water untilfurther analysis. The membrane pieces were rinsed with 100–200�l of 40% acetonitrile and 50 mM NH4HCO3, pH 7.5 and digestedwith 5–10 �l of trypsin (Promega; 15 ng/�l in 80% acetonitrile and50 mM NH4HCO3, pH 7.5) for at least 4 h at 37 °C. The reactionwas stopped by adding a 2-fold volume of 0.5% trifluoroacetic acid.The water-acetonitrile solution containing hydrophilic andslightly hydrophobic peptides was removed to a clean Eppendorftube, and an aliquot (1–2 �l) was later used to confirm the proteinidentity by MALDI-TOF MS analysis. The strongly hydrophobicpeptides were further extracted from the membrane pieces with5–10 �l of hexafluoroisopropanol for 1 h. The water-acetonitrileand hexafluoroisopropanol extracts were combined with themembrane pieces and subjected to chloroform (or chloroform/methanol) extraction. The organic phase enriched with S-acylatedpeptides was analyzed by MALDI-TOF MS.

MALDI-TOF MS Analysis—Sample aliquots (1 �l) were mixedon a steel target with an equal volume of a 2,5-dihydroxybenzoicacid (Aldrich) in 30% acetonitrile and 0.5% trifluoroacetic acidwater solution (10 mg/ml). Most mass spectra were recorded on anUltraflextreme MALDI TOF-TOF mass spectrometer (BrukerDaltonik, Germany) equipped with a 355 nm neodymium laser.The MH� molecular ions were measured in reflector mode; theaccuracy of mass peak measuring was within 0.01%. The propor-tion of stearate/palmitate was calculated from the MS peak inten-sities using the method developed earlier (35).

Amino Acid Frequency Analysis—The influenza A virus HAamino acid sequences were extracted from the Influenza ResearchDatabase in April 2014 (33,254 entries of H1–H17 subtypes,excluding laboratory strains, with complete HA sequence).Among them, there were 17,311 unique sequences chosen formultiple alignments. The MAFFT program (36) working with verylarge samplings was used to align sequences within the subtype,whereas the MUSCLE server (37) was further used to align thesubtype consensus sequences. The consensus sequence alignmentwas visualized using JalView. The frequency value F for every con-sensus amino acid X in position i in the multiple alignment wascalculated as F � (#X/N) � 100% where #X is the number ofsequences having amino acid residue X in position i, and N is thetotal number of sequences aligned.

RESULTS

Rationale for Mutagenesis in the Membrane-anchoringDomain of HA—In principle, the differential acylation patternfor HA might be influenced by the location of the acylated cys-teine relative to the transmembrane span or (but mutually notexclusive) by the presence of specific amino acids near the

Differential Acylation of Influenza HA

34980 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 50 • DECEMBER 12, 2014

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 4: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

acylation site. We first performed a bioinformatics comparisonof all HA sequences of influenza A virus present in the NCBIdatabase to reveal conserved residues in the TMR and cytoplas-mic tail that might be part of a complex acylation signal (Fig. 1).We mutated residues in H1 subtype HA from the WSN strainthat might interact with the active center of a DHHC protein,i.e. located in the inner part of the transmembrane region or inthe cytoplasmic tail (Fig. 2).

One of the latter is the hydrophobic sequence ICI at the Cterminus where the terminal isoleucine 565 and the palmitoy-lated cysteine 564 are present in each HA variant. The adjacent

isoleucine 563 is almost completely conserved through all HAsubtypes; in some subtypes it is replaced by other large hydro-phobic amino acids. To investigate whether the acylation pat-tern of HA is determined by the C-terminal hydrophobic patch,Ile-563 was exchanged by the polar residue glutamine (I563Q)or by the hydrophobic residue leucine (I563L).

Glycine at position 557 in the cytoplasmic tail is also com-pletely conserved through all HA sequences. We exchanged itby the small residue alanine (G557A) or by the negativelycharged glutamic acid (G557E). In another mutant, the cyto-plasmic tail (including the two palmitoylated cysteines) wasdeleted by replacing the codon for glycine 557 with a stop codon(G557Stop). Position 560 located adjacent to a palmitoylatedcysteine is not conserved through all HA sequences but accom-modates only two consensus amino acids, either a glutamine oran arginine, depending on the subtype. The glutamine presentin HA of the WSN strain was replaced by glutamic acid(Q560E).

Each HA subtype (except H4) contains in the middle of itsTMR a glycine, which has a very low propensity to be present inan �-helical structure. It thus might induce a kink that presentsthe cysteine at the end of the TMR to an acyltransferase. Weexchanged glycine 547 by serine (G547S), which is better com-patible with an �-helical structure.

To determine whether the location of a cysteine relative tothe transmembrane span is the main determinant for attach-ment of stearate, we shifted the cysteine at the end of the TMRto a cytoplasmic location. To do so, cysteine 554 was replacedby a serine, and leucine 559 was exchanged by cysteine (C554S/L559C). The resulting mutant thus contains an identical loca-tion of cysteines as in H8 and H9 subtype HAs from naturallyoccurring avian, equine, swine, and human virus strains.

Having verified the mutation by sequencing, the mutant HAplasmid together with seven plasmids encoding the other viralproteins were transfected into HEK 293T cells, and 3 days laterthe supernatant was tested for virus particle release by a plaqueassay. For two mutants, G557Stop and I563Q, we never rescuedinfectious virus particles, although control experiments usingwild type HA done in parallel showed that transfection wassuccessful. Thus, the mutation might have destroyed one of theessential activities of HA, i.e. virus assembly and buddingand/or membrane fusion during virus entry. All the othermutations in HA were not lethal: the nucleotide sequence wasstable during several passages of replication, but viral titerstested in a growth curve were reduced by 1 or 2 orders of mag-nitude (data not shown).

Analyzing S-Acylation of HA with Lethal Mutations—Thelethal replication defect of viruses containing the HA mutantsI563Q and G557Stop might be due to a complete defect inacylation of HA, a modification essential for virus replication.To address whether the cytoplasmic tail is an essential part ofan acylation signal, we expressed mutant HA with a vacciniavirus T7 system in CV1 cells and analyzed acylation by meta-bolic labeling with [3H]palmitate (Fig. 3). Labeling with[35S]methionine/cysteine done in parallel, immunoprecipita-tion, SDS-PAGE under non-reducing conditions, and fluorog-raphy showed two HA bands, which are likely to represent themannose-rich precursor with higher SDS-PAGE mobility and

FIGURE 1. Consensus sequence of the TMR and cytoplasmic tail for eachHA subtype. Acylation sites, the stearoylated cysteine at the end of the TMR,and the two palmitoylated cysteines in the cytoplasmic tail are highlighted inyellow. The C terminus contains the hydrophobic patch ICI where the C-ter-minal isoleucine is completely conserved (highlighted in dark blue) and theother isoleucine (highlighted in light blue) is in some subtypes replaced by aphenylalanine or valine. Also completely conserved is a glycine in the cyto-plasmic tail (highlighted in purple). Another position in the cytoplasmic tailcontains either a glutamine (dark green) or an arginine (light green). The loca-tion of the glycine(s) in the middle of the TMR present in all subtypes (exceptH4) is highlighted in pink. Above the horizontal bar are group 1 HAs; below aregroup 2 HAs (49).

FIGURE 2. Mutations introduced into the TMR and cytoplasmic tail of HA.The amino acid sequence at the inner part of the transmembrane region andcytoplasmic tail of H1 subtype HA from the WSN strain is shown. Acylatedcysteine residues are highlighted in gray; amino acid exchanges are in bold.The bars above the sequence depict the frequency of individual amino acidsthrough all HA sequences present in the NCBI database determined asdescribed under “Experimental Procedures.” � or � indicates whether infec-tious virus could be rescued by transfection of cells.

Differential Acylation of Influenza HA

DECEMBER 12, 2014 • VOLUME 289 • NUMBER 50 JOURNAL OF BIOLOGICAL CHEMISTRY 34981

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 5: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

the slower migrating product with complex glycans that hasbeen described for HA for WSN (38). The intensity of the upperand lower bands differed between wild type HA and themutants; especially HA with deleted tail exhibits more of theprecursor relative to the mature form, indicating that its intra-cellular transport is retarded. The fluorogram of the [3H]palmi-tate-labeled samples revealed that each HA mutant is labeledbut with varying intensity. Densitometric quantification of[35S]methionine/cysteine- and [3H]palmitate-labeled bandsrevealed no meaningful reduction in palmitoylation. The (non-lethal) HA mutant Q560E and the mutant I563Q are acylatedwith only slightly reduced efficiency (�90%) relative to wildtype HA (100%). Acylation of G557Stop in which two acylatedcysteines are deleted is reduced to �60%, suggesting that theone remaining acylation site at the end of the TMR is efficientlyused. Thus, the mutations introduced into HA did not preventacylation, but subtle differences in the occupancy of individualacylation sites cannot be excluded.

Establishment of a New Sample Preparation Procedure forMass Spectrometry—In our previous protocol of sample prepa-ration for MS analysis, the HA ectodomain was removed bydigestion of virus particles with bromelain, shaved particleswere pelleted, and the membrane-anchoring fragments of HAwere extracted to the chloroform phase and analyzed byMALDI-TOF MS. To remove impurities that might affect pro-teolytic cleavage of HA, virus particles were purified from theallantoic fluid of embryonated chicken eggs by sucrose gradientpurification. Because the viral mutants were compromised ingrowth, we thought to develop an alternative procedure thatdoes not require the materially demanding step of gradientpurification and can thus be performed with viruses pelletedfrom the supernatant of MDCK cells. In the new approach,virus particles are separated by non-reducing SDS-PAGE, andproteins are blotted onto PVDF membranes and visualized byPonceau S staining. The HA band is cut from the membraneand digested with trypsin, and the resulting peptides are elutedand analyzed by MALDI-TOF MS (Fig. 4A).

To test whether the new approach yields reliable results, wecompared both procedures with the same virus preparation,WSN wild type virus grown in MDCK cells. Each procedure

utilizes a different protease, and therefore different peptideswere observed in the MS spectra (Fig. 4B). According to itsspecificity, trypsin can cleave the polypeptide chain after argi-nine or lysine. We found that it cuts HA mainly after arginine513 located 15 amino acid residues above the TMR, resulting ina peptide that matches to the complete membrane-anchoringplus linker region spanning amino acids 514 –565. Two othercleavage sites after lysines 515 and 520 are also present in lowerproportion (Fig. 4C). Bromelain possesses a wider substratespecificity compared with trypsin, but only glycine 518 andlysine 520 located in the linker region connecting the TMR withthe well structured HA ectodomain are spatially accessible forthe enzyme. Thus, the resulting peptides are shorter, spanningamino acids 519 –565 and 521–565, respectively. Each of thementioned peptides is present as two main peaks representingtriple palmitoylated peptide and a peptide containing twopalmitates and one stearate. A very small third peak represent-ing a peptide containing one palmitate and two stearates alsoappears sometimes. Calculation of the amount of stearaterevealed very similar results: 20.1% for the new blotting proce-dure and 21.1% for the previous extraction method. Thus, thenew method is reliable and able to detect acylated peptidesusing �5–10 �g of protein, i.e. a Ponceau S-stained band.

Surprisingly, however, the stearate content from HA of theWSN virus was much higher than the amount previously deter-mined for the closely related PR8 virus (12.6%; Ref. 24) thatcontains a very similar C-terminal amino acid sequence.Because the viruses were grown in MDCK cells and embryo-nated eggs, respectively, we hypothesized that the host cellsmight affect the acylation pattern of HA.

Comparison of Acylation Patterns of HA Synthesized in Avianand Mammalian Cells—All our previous MS analyses were per-formed with virus strains purified from an avian host, embryo-nated chicken eggs. In this system, HAs from human viruses haveless stearate (12–18%) compared with HAs from avian viruses(18–35%; Refs. 24 and 27). To analyze whether HA from humanviruses contains more stearate if grown in mammalian cells, weperformed MS analysis with WSN using a variety of samples. Wecompared recombinant WSN (generated from transfectedMDCK cells and then further amplified either in eggs or in MDCKcells) with a natural WSN virus strain amplified in eggs or inMDCK cells using both the previous extraction method and thenew blotting procedure. In each case, WSN virus grown in eggs hasless HA-bound stearate (8–12%) compared with WSN grown inMDCK cells (20.1–22.9%). Furthermore, to corroborate the sur-prising finding that avian cells apparently attach less stearate toHA compared with mammalian cells, we also analyzed WSNgrown in CEFs, primary cells prepared from chicken embryos, andWSN grown in DF-1 cells, a cell line derived from CEFs. Theamount of stearate attached to HA from CEF-derived WSN wasidentical (7.7%) to that attached to HA from egg-derived WSN(7.9%). DF-1 cells attach somewhat more stearate to HA (10.4%)but much less than MDCK cells (22.9%; results are summarized inTable 1). Thus, neither the sample preparation for MALDI-TOFMS nor the origin of the virus (from plasmid or from virus strain)significantly affected the result, indicating that mammalian cellsattach more stearate to human viruses compared with cells ofavian origin.

FIGURE 3. Expression and [3H]palmitoylation of non-functional HAmutants. HA mutants were expressed with a vaccinia virus T7 system in CHOcells. Transfected cells were labeled for 3 h with [35S]-methionine/cysteineand [3H]palmitate, respectively. HA was immunoprecipitated from cellextracts and subjected to non-reducing SDS-PAGE and fluorography. Thelower HA band (HA*) is a mannose-rich precursor, and the upper HA band isthe mature product (38). Densitometric quantification of both [35S]methio-nine/cysteine-labeled bands and the [3H]palmitate band revealed a 3H to 35Sratio of 90% for Q560E and 86% for I563Q relative to wild type HA (100%). TheG557Stop mutant has a 3H to 35S ratio of 62%, but two acylation sites aredeleted in this construct. mock, untransfected cells.

Differential Acylation of Influenza HA

34982 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 50 • DECEMBER 12, 2014

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 6: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

We then analyzed HA from the PR8 strain, which is of inter-est because it is used as a backbone to make seasonal flu vac-cines; i.e. the glycoproteins of circulating human strains arereassorted into PR8, and the resulting virus strain is amplified inembryonated eggs for vaccine production. Identical to HA ofWSN, we observed a tryptic peptide that matches to the com-plete membrane-anchoring region spanning amino acids 514 –565 (Fig. 5). Calculation of the stearate amount in the two peaksrevealed low stearate content in HA if the virus was grown ineggs (8.2%) compared with virus obtained from MDCK cells(21.2%). A low stearate amount was also previously determined

with egg-grown PR8 virus using the previous extractionmethod (24). We conclude that HA from human strains isincompletely stearoylated if the virus is amplified in avian cells.However, no underacylated peptides were observed, indicatingthat the stearoylation site at the end of the TMR is occupied bypalmitate. This was earlier confirmed by MALDI-TOF-TOFfragmentation analysis (35).

Exchange of Amino Acids Modestly Affects the AcylationPattern—Next we determined the fatty acid pattern of mutantHAs from recombinant WSN viruses grown in MDCK cellsusing the blotting procedure. Because the error rate of the viral

FIGURE 4. Establishing a new method for sample preparation prior to MS. A, SDS-PAGE analysis under non-reducing conditions and Coomassie staining ofA/WSN/33 (H1N1) virions grown in MDCK cells before (�) and after (�) digestion with bromelain and pelleting (100,000 � g, 1.5 h, 4 °C) of particles. Thepositions of viral proteins (HA, NP, and M1) and molecular mass markers in kDa are indicated. I, the new protocol includes electroblotting of undigested virusparticles onto a PVDF membrane, “on-membrane” digestion of HA with trypsin, peptide elution, and MS analysis. II, the previous protocol is based on theproteolytic treatment of purified virions with bromelain followed by chloroform/methanol (Ch/Met) extraction of the anchoring peptides and their analysiswith MS. B, MALDI-TOF MS analysis of S-acylated HA peptides obtained by methods I and II. Mass spectra obtained in reflector mode are depicted. Indicated arethe m/z values of the peaks, the first and last amino acid residues of the corresponding peptides, and the number and type of HA-bound fatty acids. Amountsof HA-bound stearate (mean percentage �S.D.) calculated from three to five spectra obtained for every of three blotting repeats (I; peaks of peptide HA(514 –565) were taken for calculation) or from one bromelain digestion (II; both peptides HA(516 –565) and HA(521–565) were used for calculation) are indicated.Minor peaks occasionally found between the peptides containing 3Pal and 2Pal1Stear and between 2Pal1Stear and 1Pal2Stear, respectively, probably resultfrom methylation in acid medium (�14) or Met oxidation (�16). Note that non- or monoacylated HA peptides (peaks with m/z values around 4845 and 5083,respectively) were not detected in the spectra. C, amino acid sequence of the HA C-terminal region indicating sites of cleavage by trypsin (I) or bromelain (II).The arrow lengths approximately correspond to the respective peak heights in the spectra. Cysteines as potential acylation sites are enlarged, and the TMR isunderlined.

TABLE 1Comparison of amount of HA-bound stearate in virus grown in avian and mammalian cells determined by MALDI-TOF MSResults are represented as mean � S.D. The remainder is palmitate.

WSN recombinant virus WSN natural virus strain PR8 natural virus strainEgga MDCKa CEFa DF-1a Egga Eggsb MDCKa Egga Eggsb,c MDCKa

7.9 � 4.7 22.9 � 1.2 7.7 � 0.7 10.4 � 1.5 7.6 � 1.5 12.1 � 0.8 20.1 � 0.9 8.2 � 0.4 12.6 � 1.7 21.2 � 0.4a New blotting protocol.b Previous extraction protocol.c Published in Ref. 24.

Differential Acylation of Influenza HA

DECEMBER 12, 2014 • VOLUME 289 • NUMBER 50 JOURNAL OF BIOLOGICAL CHEMISTRY 34983

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 7: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

polymerase is high and virus with nucleotide reversions to thewild type sequence rapidly outgrows the mutant virus, wesequenced the genomic RNA encoding the C terminus of HA,and only virus preparations with the desired sequence wereanalyzed by MALDI-TOF MS. Similar to the wild type HA,trypsin cleaves the polypeptide chain after arginine 514 but alsoafter arginine 516 such that two peptides appear in the spectrathat encompass the cleavage site until the C terminus of HA(Fig. 6). The observed peptide masses are also consistent withthe expected sequences shown in Fig. 2, indicating that noreversion to wild type (or any other amino acid) occurred.

Each of the mentioned peptides is present as two main peaksrepresenting triple palmitoylated peptide and a peptide con-taining two palmitates and one stearate. However, the intensi-ties of the peaks clearly differed between the HA mutants, indi-cating that the introduced amino acid replacements changedthe acylation pattern. We calculated the stearate content forthis and usually one other virus preparation; the results aregraphically shown in Fig. 9.

A reduction in the stearate content was observed only forG557E, which contains 15% of the longer acyl chain in compar-ison with 23% stearate calculated for wild type HA. Because theconservative replacement of the completely conserved glycine557 by an alanine has no significant effect (24% stearate), it israther the insertion of the acidic glutamic acid that is responsi-ble for the observed stearate reduction in G557E. Exchange ofglutamine by glutamic acid in position 560 caused a very minorincrease in the stearate proportion (26%), indicating that theinsertion of a negatively charged residue into the cytoplasmictail does not reduce stearoylation per se. However, the conser-vative replacement I563L at the hydrophobic C terminus of HAsignificantly raised the stearate proportion to 31%. The largestincrease in the stearate content to 33% was observed with theHA mutant that contains a serine instead of the glycine locatedin the middle of the TMR.

We also analyzed one of the HA mutants (G557A) with egg-grown virus using the previous, better established procedurefor sample preparation (Fig. 7). Around 12% stearate was deter-mined; this is identical to the wild type HA analyzed with thesame procedure but half as much as was calculated for the samemutant virus grown in MDCK cells. This result reinforces ournotion that avian cells attach less stearate to the HA of humanviruses compared with mammalian cells. In addition, non- orunderacylated peptides were not detected, suggesting that themutation did not alter the stoichiometry of S-acylation. In sum-mary, point mutations introduced into the TMR or cytoplasmictail of HA modestly affected the fatty acid pattern of HA but didnot abolish stearoylation or S-acylation per se.

Shifting a Cysteine from the TMR to the Cytoplasm EliminatesAttachment of Stearate—Finally, we determined the fatty acidpattern of the C554S/L559C mutant grown in MDCK cells; inthis mutant the usually stearoylated transmembrane cysteinewas shifted to a cytoplasmic location. Only very small peaksrepresenting peptides with two palmitates and one stearate aredetectable; the by far predominant peptide contains threepalmitates (Figs. 8 and 9). Calculation of the stearate contentrevealed that it is reduced to 3.3%.

We also analyzed the fatty acid pattern of an H9 subtype HAfrom a teal virus grown in embryonated eggs that has the samespacing of cysteines embedded in a different amino acid context(SCRCNICI (H9) versus SCQCRICI (H1)). Despite these differ-ences in the amino acid sequence, the same small peak intensi-ties were detected for peptides with two palmitates and onestearate compared with those with three palmitates in the spec-tra of H9 and H1 HAs. Thus, the location of an acylated cysteinerelative to the end of the transmembrane span is the decisivefactor for the attachment of palmitate versus stearate as dem-onstrated both for an egg-grown avian virus and for a humanvirus from MDCK cells.

FIGURE 5. MS result from PR8 virus grown in avian and mammalian cells. A, non-reducing SDS-PAGE and Coomassie staining of A/Puerto Rico/34 (H1N1)virions grown in MDCKII cells or in embryonated eggs. The positions of viral proteins (HA, NP, and M1) and molecular mass markers in kDa are indicated. Thestrong band below NP is probably albumin from the serum used to grow virus in cell culture. HA was analyzed with the new blotting-MS protocol. B, MALDI-TOFMS analysis. Mass spectra obtained in reflector mode are depicted. Indicated are the m/z values of the peaks, the first and last amino acid residues of thecorresponding peptides, and the number and type of HA-bound fatty acids. For simplicity, additional minor peaks producing the peptides HA(516 –565) andHA(521–565) with lower m/z values are neither shown nor used for stearate calculation. The calculated stearate content found for the HA(514 –565) peptidesfrom several spectra is listed in Table 1. C, amino acid sequence of the HA C-terminal region indicating the prevailing site of cleavage by trypsin. Cysteines aspotential acylation sites are enlarged, and the TMR is underlined.

Differential Acylation of Influenza HA

34984 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 50 • DECEMBER 12, 2014

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 8: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

DISCUSSION

In this study, we used reverse genetics and mass spectrome-try to determine the molecular signals that determine site-spe-cific attachment of palmitate and stearate to HA of influenzavirus. We first developed a new sample preparation procedurefor MS analysis of acylated proteins that uses non-reducingSDS-PAGE of a virus preparation, blotting to PVDF mem-branes, identification of HA on the membrane by Ponceau Sstaining, in situ proteolysis, and elution of peptides from themembrane by organic solvents prior to MALDI-TOF MS. Wehave shown that the procedure produces the same result as theprevious approach using proteolysis of gradient-purified virusparticles (Fig. 4). The variation of the results between differentvirus preparations was also low (1– 6%), albeit somewhat higher

compared with the previous procedure (1–3%). Both protocols(proteolytic digestion of virions/MS or protein blotting/MS)demonstrated similar �1% (or less) variation when repeated forthe same virus preparation. Nevertheless, the new approach mightrequire only �10 �g of acylated protein to calculate the stearate/palmitate ratio and probably less just to detect the acylated pep-tides and is thus quite suitable to identify acylation patterns ofabundantly expressed cellular membrane proteins. Purificationof the protein is not even required as long as the protein ofinterest can be identified in a protein mixture as an individual(well represented) band by Ponceau S staining.

Using both the new and previous procedures, we found thatHA from two human H1N1 strains (WSN and PR8) containsmore stearate (20 –23%) if grown in mammalian MDCK cells

FIGURE 6. MS results from WSN mutants with exchange of amino acids. A, non-reducing SDS-PAGE and Coomassie staining of A/WSN/33(H1N1) recombi-nant virus particles grown in MDCKII cells. The mutation introduced into HA, the positions of viral proteins (HA, NP, and M1), and molecular mass markers in kDaare indicated. HA was analyzed with the new blotting-MS protocol. B, MALDI-TOF MS analysis. Mass spectra obtained in reflector mode are depicted. Indicatedare the m/z values of the peaks, the first and last amino acid residues of the corresponding peptides, and the number and type of HA-bound fatty acids. A thirdpeak with m/z values around 5557 corresponding to the triple acylated peptide HA(521–565) was also observed but is not shown because it overlaps withthe trypsin autolysis peak at 5561 m/z. The stearate amount calculated for the peptides HA(516 –565) and/or HA(514 –565) of each mutant is depicted in Fig. 9.The m/z region corresponding to non- and underacylated peptides is not included in the spectra, but such peptides were never observed. C, amino acidsequence of the HA C-terminal region indicating the amino acid exchange and the prevailing site of cleavage by trypsin. Cysteines as potential acylation sitesare enlarged, and the TMR is underlined.

Differential Acylation of Influenza HA

DECEMBER 12, 2014 • VOLUME 289 • NUMBER 50 JOURNAL OF BIOLOGICAL CHEMISTRY 34985

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 9: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

compared with the same virus amplified in avian cells (8 –12%;Fig. 5 and Table 1). Together with the observation that HA fromavian viruses already contains a high proportion of stearate(22–35%) when the virus is grown in eggs (24, 27), we speculatethat viruses have adapted to their natural host cells such thatthey receive a high stearate proportion. However, before anyfirm conclusions can be drawn, more experiments with avian,porcine, equine, and human influenza viruses grown in naturaland non-natural host cells should be performed. Nevertheless,this observation already indicates that there are cell type-spe-cific differences in the acylation pattern of HA and thus prob-ably also in cellular proteins. In addition, our results add to thevery recent finding that influenza virions produced by mamma-lian and avian cells have distinct protein compositions, indicat-ing that the host cell influences the architecture of virus parti-cles (39).

We also showed that none of the introduced mutations pre-vented S-acylation of HA. The deleterious mutations in HA thatdid not allow generating infectious virus particles, i.e. I563Qand G557Stop, are acylated as efficiently as wild type HA as

deduced from metabolic labeling with [3H]palmitate (Fig. 3).Thus, the functional defect in HA introduced by the mutation isnot due to a significant blockade of acylation. In the MS spectrafrom HA mutants with which we could generate infectious viri-ons, we never detected peaks that correspond to non-acylatedor underacylated peptides (Figs. 4 – 8). Thus, the conserved gly-cine residues located in the cytoplasmic tail (Gly-557) and inthe TMR (Gly-547) and Gln-560 in the cytoplasmic tail areapparently essential for acylation of HA. It is even possible toshift a cysteine from the TMR to a cytosolic location withoutcompromising attachment of fatty acids at any of the threeacylation sites.

However, a word of caution regarding the quantitative nature ofMS data is necessary. Although in principle we could detect non-and underacylated peptides with our MALDI-TOF setting (35), wepresently cannot exclude that non- or underacylated peptides con-taining exchange of crucial amino acids “do not fly” or are lostduring sample preparation. Thus, we cannot rule out that somepeptides are underacylated, i.e. that the mutations reduced theefficiency of acylation at certain sites. Nevertheless, it is fair to

FIGURE 7. MS results from WSN wild type and G557A mutant grown in avian cells. A, non-reducing SDS-PAGE and Coomassie staining of A/WSN/33(H1N1)virions (control) and WSN G557A recombinant viral particles grown in eggs before (�) and after (�) digestion with bromelain and pelleting (100,000 � g, 1.5 h,4 °C). The bromelain-digested viral particles were further analyzed with the previous extraction-MS protocol. The positions of viral proteins (HA, NP, and M1)and molecular mass markers in kDa are indicated. B, MALDI-TOF MS analysis. Mass spectra obtained in reflector mode are depicted. Indicated are the m/z valuesof the peaks, the first and last amino acid residues of the corresponding peptides, and the number and type of HA-bound fatty acids. Indicated is the percentageof HA-bound stearate (mean � S.D.) calculated from four to six spectra obtained for each of one to three digestions performed for three (control) or one(G557A) virus preparations. Note that non- or monoacylated HA peptides (peaks with m/z values around 4845 and 5083, respectively) were not detected in thespectra, and only very tiny peaks of doubly acylated peptides (m/z around 5321) were observed. C, amino acid sequence of the HA C-terminal region indicatingthe amino acid exchange and the site of cleavage by bromelain. The arrow lengths approximately correspond to the respective peak heights in the spectra.Cysteines as potential acylation sites are enlarged, and the TMR is underlined.

Differential Acylation of Influenza HA

34986 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 50 • DECEMBER 12, 2014

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 10: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

conclude that a putative acyltransferase does not need to recognizeand interact with amino acids in the vicinity of acylation sites as anessential step of its catalytic cycle. Most likely, a reactive cysteine,i.e. reduced and deprotonated, that is accessible to the active site ofan enzyme, i.e. exposed near the cytoplasmic face of the mem-brane, is sufficient for acylation to occur (40).

We also conclude that the location of a cysteine relative tothe end of the TMR is the decisive factor for attachment ofstearate. Shifting the cysteine to a cytoplasmic location reducedattachment of the longer chain fatty acid to 3%. The same pro-portion of stearate was found in the HA of a natural H9 subtypevirus strain (Fig. 8), in the HA of H7 subtype if the TMR cysteinewas exchanged for a serine (24), and in the HA of influenza Bvirus that evolutionarily lacks a cysteine residue at the end ofthe TMR (24). It is now obvious that the length of the cytoplas-mic tail originally proposed to be important for stearoylation(41) plays no role at all because the fusion protein of Newcastledisease virus is stearoylated at a TMR cysteine despite contain-ing a long (27-amino acid) cytoplasmic tail (26).

Nevertheless, an exchange of amino acids subtly influencesthe proportion of stearate attached to HA (Fig. 6). The mostprominent effect was calculated for the mutant G547S, whichshowed an increase in the stearate proportion from 23 (wildtype HA) to 33%. We hypothesize that the exchange of theconserved glycine in the TMR by a serine, which is more com-patible with an �-helical conformation, straightens the trans-membrane span and places the TMR cysteine in a better posi-tion for stearoylation. Alternatively, it was proposed thatglycine residues in the transmembrane region are orientedtoward the helix-helix interface to mediate interactions (42).Indeed, molecular modeling of the closely related H6 subtype

FIGURE 8. MS results from a WSN mutant and a natural H9 subtype HA with shifted cysteine. The A/WSN/33 (H1N1) recombinant virus mutant (C554S/L559C)was grown in MDCK cells and analyzed using the blotting approach, whereas the A/Teal/Primorie3631/02 (H9N2) virus was grown in embryonated chicken eggs andanalyzed using the previous protocol based on bromelain digestion of virions. Mass spectra obtained in reflector mode are depicted. Indicated are the m/z values ofthe peaks, the first and last amino acid residues of the corresponding peptides, and the number and type of HA-bound fatty acids. The stearate content was calculatedas 3.3% for the H1 subtype mutant and 4.5% for H9 subtype HA. The peaks of both H1 subtype HA(516–565) and HA(514–565) and H9 subtype HA(518–560) andHA(516–560) were used for calculations. Depicted below the spectra are the C-terminal amino acid sequences of H1 and H9 subtype HAs indicating the cleavage sitesby trypsin and bromelain, respectively. Cysteines as potential acylation sites are enlarged, and the TMR is underlined.

FIGURE 9. Graphical summary of MS results with recombinant WSN virusfrom MDCK cells. Stearate content was calculated from one (I563L andC554S/L559C), two (G557E, G557A, Q560E, and G547S) or three (WT) viruspreparations. For each virus preparation, at least two blotting repeats wereperformed, and at least four mass spectra were recorded. The remainder ispalmitate; the results are represented as mean percentage �S.D. (error bars).

Differential Acylation of Influenza HA

DECEMBER 12, 2014 • VOLUME 289 • NUMBER 50 JOURNAL OF BIOLOGICAL CHEMISTRY 34987

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 11: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

HA implies that this is the case for the HA TMR (27). Substitu-tion of glycine by serine might alter these interactions and/orrotate one TMR helix against the other. In all these cases, thecysteine at the end of the TMR might become better accessibleby a putative stearate-specific acyltransferase.

The conservative substitution of isoleucine with leucine at thehydrophobic patch at the C terminus of HA (and thus at a distanceof nine amino acids from the TMR cysteine) increased the stearateproportion to 31%. Because of limited sample material, we couldnot identify the fatty acid pattern for each acylation site withMS/MS sequencing as we did in our previous analysis. However, inall our previous analyses, we never observed attachment of stearateat sites other than the TMR cysteine. Thus, it is likely that addi-tional stearate (relative to the wild type HA) is also attached hereand not at the C-terminal cysteine 564. One might speculate thatthe hydrophobic patch at the C terminus of the cytoplasmic tailinteracts with the TMR, thereby contributing to a subtle increaseof the stearate content at cysteine 554.

A reduction in the stearate content to 15% was observed in themutant G557E. This is not due to replacement of the completelyconserved, cytosolic glycine residue because its exchange by analanine had no effect on the stearate content. It is also not causedby the presence of glutamic acid per se because in the mutantQ560E a slight increase in the stearate content was noticed. Wesuppose that insertion of a negatively charged residue at the mem-brane-cytosol interface pulls the cysteine out of the membrane andexposes it to the cytosol such that it is in a less favorable location forstearate attachment. Because the TMR of HA is very long (27amino acids), it has a wide range to move perpendicular to theplane of the membrane, especially at the intracellular site of acyla-tion (the late endoplasmic reticulum or early Golgi region) wherethe membrane bilayer is supposed to be thinner. In summary, con-servative as well as drastic exchanges of amino acids in variousregions at the C terminus of HA modestly alter the stearateproportion in a rather unpredictable manner. We hypothe-size that the mutations alter the location of the cysteinewithin the transition region between the membrane bilayerand the cytosol, thereby affecting its accessibility for a puta-tive stearate-specific acyltransferase.

Our last conclusion alludes to the mechanism of S-acylationof HA, i.e. whether attachment of palmitate and/or stearaterequires an enzyme or whether only the presence of the lipiddonor, i.e. Pal-CoA or Stear-CoA, is sufficient. Because acyl-CoAs are sequestered with very high affinity (�0.1 nM) by theacyl-CoA-binding protein (43), their cytosolic concentration ispresumably too low to facilitate spontaneous acylation. How-ever, acyl-CoA partitions into lipid bilayers with a high parti-tion constant (1–5 � 105/M) by insertion of its acyl chain. Usingatomic force microscopy and supported lipid bilayers, it wasfound that acyl-CoA forms lateral aggregates in the plane of thebilayer (44) that might represent the lipid donor for non-enzy-matic S-acylation if such clusters exist in living cells. One mightimagine that a fatty acid is spontaneously transferred to a pro-tein if acyl-CoA encounters a cysteine at the end of the TMR ofa protein in transit along the exocytic pathway.

However, such a non-enzymatic reaction would not show anypreference for the attachment of a particular fatty acid to a certainsite but should rather reflect the concentration of individual acyl-

CoAs at the intracellular site of acylation, the membranes of thelate endoplasmic reticulum or early Golgi (19). Although very littleis known about the distribution of acyl-CoAs in cellular mem-branes, there is no indication that the concentrations of Pal-CoAand Stear-CoA grossly differ in the endoplasmic reticulum/Golgibecause both are also used for the synthesis of phospholipids thatcontain palmitate and stearate as the major type of fatty acids.

In addition, the cell type-specific heterogeneity we found in theacylation pattern of HA is also difficult to reconcile with a non-enzymatic mechanism of S-acylation. Moreover, although insectcells are able to acylate HA when expressed with the baculovirussystem (45), attachment of stearate was observed neither for HAnor for endogenous insect proteins (46). Thus, stearoylation isapparently an evolutionary achievement of higher cells, and theresponsible enzyme(s) might have then acquired slightly differentsubstrate specificities in mammalian and avian cells.

In sum, it is more likely that (at least) two enzymes exist thatdiffer in their acyl-CoA specificities as recently demonstrated forDHHC2 and -3 (47). One can imagine that the enzyme responsiblefor palmitoylation recognizes cysteine exposed to the hydrophilicmilieu of the cytosol. In contrast, the active site of a DHHC proteinwith a strong preference for Stear-CoA might have a certain affin-ity for the unique biophysical environment of the border regionbetween the hydrophobic and hydrophilic parts of the lipid bilayer.However, it cannot penetrate deeply into the lipid bilayer becauseH7 and H10 subtype HAs contain one cysteine and H3, H4, andH14 subtype HAs contain two cysteines in the middle of theirtransmembrane regions (Fig. 1) that are not acylated (4, 27). Iden-tification of the DHHC proteins that acylate HA is a prime aim ofour future investigations. Albeit 23 or 24 different DHHC proteinswith distinct, partly overlapping substrate specificities are encodedby the mammalian genome (17, 18, 48), only a few might catalyzeacylation of HA. These DHHC proteins are promising drug can-didates because their inhibition might not compromise acylationof most cellular proteins.

Acknowledgments—We are grateful to Dr. S. G. Markushin and E. A.Kropotkina for providing us natural WSN and PR8 virus strain prep-arations. The MALDI MS facility was made available in the frame-work of Moscow State University Development Program PNG 5.13.

REFERENCES1. Skehel, J. J., and Wiley, D. C. (2000) Receptor binding and membrane

fusion in virus entry: the influenza hemagglutinin. Annu. Rev. Biochem.69, 531–569

2. Naeve, C. W., and Williams, D. (1990) Fatty acids on the A/Japan/305/57influenza virus hemagglutinin have a role in membrane fusion. EMBO J. 9,3857–3866

3. Steinhauer, D. A., Wharton, S. A., Wiley, D. C., and Skehel, J. J. (1991)Deacylation of the hemagglutinin of influenza A/Aichi/2/68 has no effecton membrane fusion properties. Virology 184, 445– 448

4. Veit, M., Kretzschmar, E., Kuroda, K., Garten, W., Schmidt, M. F., Klenk,H. D., and Rott, R. (1991) Site-specific mutagenesis identifies three cys-teine residues in the cytoplasmic tail as acylation sites of influenza virushemagglutinin. J. Virol. 65, 2491–2500

5. Chen, B. J., Takeda, M., and Lamb, R. A. (2005) Influenza virus hemagglu-tinin (H3 subtype) requires palmitoylation of its cytoplasmic tail for as-sembly: M1 proteins of two subtypes differ in their ability to supportassembly. J. Virol. 79, 13673–13684

6. Wagner, R., Herwig, A., Azzouz, N., and Klenk, H. D. (2005) Acylation-

Differential Acylation of Influenza HA

34988 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 50 • DECEMBER 12, 2014

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 12: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

mediated membrane anchoring of avian influenza virus hemagglutinin isessential for fusion pore formation and virus infectivity. J. Virol. 79,6449 – 6458

7. Zurcher, T., Luo, G., and Palese, P. (1994) Mutations at palmitylation sitesof the influenza virus hemagglutinin affect virus formation. J. Virol. 68,5748 –5754

8. Engel, S., Scolari, S., Thaa, B., Krebs, N., Korte, T., Herrmann, A., and Veit,M. (2010) FLIM-FRET and FRAP reveal association of influenza virushaemagglutinin with membrane rafts. Biochem. J. 425, 567–573

9. Levental, I., Grzybek, M., and Simons, K. (2010) Greasing their way: lipidmodifications determine protein association with membrane rafts. Bio-chemistry 49, 6305– 6316

10. Melkonian, K. A., Ostermeyer, A. G., Chen, J. Z., Roth, M. G., and Brown,D. A. (1999) Role of lipid modifications in targeting proteins to detergent-resistant membrane rafts. Many raft proteins are acylated, while few areprenylated. J. Biol. Chem. 274, 3910 –3917

11. Simons, K., and Gerl, M. J. (2010) Revitalizing membrane rafts: new toolsand insights. Nat. Rev. Mol. Cell Biol. 11, 688 – 699

12. Rossman, J. S., and Lamb, R. A. (2011) Influenza virus assembly and bud-ding. Virology 411, 229 –236

13. Melikyan, G. B., Jin, H., Lamb, R. A., and Cohen, F. S. (1997) The role of thecytoplasmic tail region of influenza virus hemagglutinin in formation andgrowth of fusion pores. Virology 235, 118 –128

14. Sakai, T., Ohuchi, R., and Ohuchi, M. (2002) Fatty acids on the A/USSR/77influenza virus hemagglutinin facilitate the transition from hemifusion tofusion pore formation. J. Virol. 76, 4603– 4611

15. Ujike, M., Nakajima, K., and Nobusawa, E. (2004) Influence of acylationsites of influenza B virus hemagglutinin on fusion pore formation anddilation. J. Virol. 78, 11536 –11543

16. Veit, M., Serebryakova, M. V., and Kordyukova, L. V. (2013) Palmitoyla-tion of influenza virus proteins. Biochem. Soc. Trans. 41, 50 –55

17. Greaves, J., and Chamberlain, L. H. (2011) DHHC palmitoyl transferases:substrate interactions and (patho)physiology. Trends Biochem. Sci. 36,245–253

18. Linder, M. E., and Deschenes, R. J. (2007) Palmitoylation: policing proteinstability and traffic. Nat. Rev. Mol. Cell Biol. 8, 74 – 84

19. Veit, M., and Schmidt, M. F. (1993) Timing of palmitoylation of influenzavirus hemagglutinin. FEBS Lett. 336, 243–247

20. Kümmel, D., Heinemann, U., and Veit, M. (2006) Unique self-palmitoyla-tion activity of the transport protein particle component Bet3: a mecha-nism required for protein stability. Proc. Natl. Acad. Sci. U.S.A. 103,12701–12706

21. Rocks, O., Gerauer, M., Vartak, N., Koch, S., Huang, Z. P., Pechlivanis, M.,Kuhlmann, J., Brunsveld, L., Chandra, A., Ellinger, B., Waldmann, H., andBastiaens, P. I. (2010) The palmitoylation machinery is a spatially organiz-ing system for peripheral membrane proteins. Cell 141, 458 – 471

22. Schmidt, M. F. (1984) The transfer of myristic and other fatty acids onlipid and viral protein acceptors in cultured cells infected with SemlikiForest and influenza virus. EMBO J. 3, 2295–2300

23. Liang, X., Lu, Y., Neubert, T. A., and Resh, M. D. (2002) Mass spectromet-ric analysis of GAP-43/neuromodulin reveals the presence of a variety offatty acylated species. J. Biol. Chem. 277, 33032–33040

24. Kordyukova, L. V., Serebryakova, M. V., Baratova, L. A., and Veit, M.(2008) S acylation of the hemagglutinin of influenza viruses: mass spec-trometry reveals site-specific attachment of stearic acid to a transmem-brane cysteine. J. Virol. 82, 9288 –9292

25. Veit, M., Herrler, G., Schmidt, M. F., Rott, R., and Klenk, H. D. (1990) Thehemagglutinating glycoproteins of influenza B and C viruses are acylatedwith different fatty acids. Virology 177, 807– 811

26. Kordyukova, L. V., Serebryakova, M. V., Baratova, L. A., and Veit, M.(2010) Site-specific attachment of palmitate or stearate to cytoplasmicversus transmembrane cysteines is a common feature of viral spike pro-teins. Virology 398, 49 –56

27. Kordyukova, L. V., Serebryakova, M. V., Polyansky, A. A., Kropotkina,E. A., Alexeevski, A. V., Veit, M., Efremov, R. G., Filippova, I. Y., andBaratova, L. A. (2011) Linker and/or transmembrane regions of influenzaA/Group-1, A/Group-2, and type B virus hemagglutinins are packed dif-

ferently within trimers. Biochim. Biophys. Acta 1808, 1843–185428. Serebryakova, M. V., Kordyukova, L. V., Semashko, T. A., Ksenofontov,

A. L., Rudneva, I. A., Kropotkina, E. A., Filippova, I. Y., Veit, M., andBaratova, L. A. (2011) Influenza virus hemagglutinin spike neck architec-tures and interaction with model enzymes evaluated by MALDI-TOFmass spectrometry and bioinformatics tools. Virus Res. 160, 294 –304

29. Serebryakova, M. V., Kordyukova, L. V., Rudneva, I. A., Kropotkina, E. A.,Veit, M., and Baratova, L. A. (2013) Mass spectrometry analysis of influ-enza virus reassortant clones does not reveal an influence of other viralproteins on S-acylation of hemagglutinin. Arch. Virol. 158, 467– 472

30. Hernandez, R., and Brown, D. T. (2010) Growth and maintenance of chickembryo fibroblasts (CEF). Curr. Protoc. Microbiol. Appendix 4, 4I

31. Neumann, G., Watanabe, T., Ito, H., Watanabe, S., Goto, H., Gao, P.,Hughes, M., Perez, D. R., Donis, R., Hoffmann, E., Hobom, G., andKawaoka, Y. (1999) Generation of influenza A viruses entirely from clonedcDNAs. Proc. Natl. Acad. Sci. U.S.A. 96, 9345–9350

32. Sugita, Y., Noda, T., Sagara, H., and Kawaoka, Y. (2011) Ultracentrifuga-tion deforms unfixed influenza A virions. J. Gen. Virol. 92, 2485–2493

33. Veit, M., Ponimaskin, E., and Schmidt, M. F. (2008) Analysis of S-acylationof proteins. Methods Mol. Biol. 446, 163–182

34. Kordyukova, L. V., Ksenofontov, A. L., Serebryakova, M. V., Ovchin-nikova, T. V., Fedorova, N. V., Ivanova, V. T., and Baratova, L. A. (2004)Influenza A hemagglutinin C-terminal anchoring peptide: identificationand mass spectrometric study. Protein Pept. Lett. 11, 385–391

35. Serebryakova, M. V., Kordyukova, L. V., Baratova, L. A., and Markushin,S. G. (2006) Mass spectrometric sequencing and acylation character anal-ysis of C-terminal anchoring segment from influenza A hemagglutinin.Eur. J. Mass Spectrom. 12, 51– 62

36. Katoh, K., and Toh, H. (2008) Recent developments in the MAFFT mul-tiple sequence alignment program. Brief. Bioinform. 9, 286 –298

37. Edgar, R. C. (2004) MUSCLE: multiple sequence alignment with highaccuracy and high throughput. Nucleic Acids Res. 32, 1792–1797

38. Rodriguez-Boulan, E., Paskiet, K. T., Salas, P. J., and Bard, E. (1984) Intra-cellular transport of influenza virus hemagglutinin to the apical surface ofMadin-Darby canine kidney cells. J. Cell Biol. 98, 308 –319

39. Hutchinson, E. C., Charles, P. D., Hester, S. S., Thomas, B., Trudgian, D.,Martínez-Alonso, M., and Fodor, E. (2014) Conserved and host-specificfeatures of influenza virion architecture. Nat. Commun. 5, 4816

40. Dietrich, L. E., and Ungermann, C. (2004) On the mechanism of proteinpalmitoylation. EMBO Rep. 5, 1053–1057

41. Veit, M., Reverey, H., and Schmidt, M. F. (1996) Cytoplasmic tail lengthinfluences fatty acid selection for acylation of viral glycoproteins. Biochem.J. 318, 163–172

42. Javadpour, M. M., Eilers, M., Groesbeek, M., and Smith, S. O. (1999) Helixpacking in polytopic membrane proteins: role of glycine in transmem-brane helix association. Biophys. J. 77, 1609 –1618

43. Faergeman, N. J., and Knudsen, J. (1997) Role of long-chain fatty acyl-CoAesters in the regulation of metabolism and in cell signalling. Biochem. J.323, 1–12

44. Cohen Simonsen, A., Bernchou Jensen, U., Faergeman, N. J., Knudsen, J.,and Mouritsen, O. G. (2003) Acyl-coenzyme A organizes laterally in mem-branes and is recognized specifically by acyl-coenzyme A binding protein.FEBS Lett. 552, 253–258

45. Kuroda, K., Veit, M., and Klenk, H. D. (1991) Retarded processing of in-fluenza virus hemagglutinin in insect cells. Virology 180, 159 –165

46. Reverey, H., Veit, M., Ponimaskin, E., and Schmidt, M. F. (1996) Differen-tial fatty acid selection during biosynthetic S-acylation of a transmem-brane protein (HEF) and other proteins in insect cells (Sf9) and in mam-malian cells (CV1). J. Biol. Chem. 271, 23607–23610

47. Jennings, B. C., and Linder, M. E. (2012) DHHC protein S-acyltransferasesuse similar ping-pong kinetic mechanisms but display different acyl-CoAspecificities. J. Biol. Chem. 287, 7236 –7245

48. Roth, A. F., Wan, J., Bailey, A. O., Sun, B., Kuchar, J. A., Green, W. N.,Phinney, B. S., Yates, J. R., 3rd, and Davis, N. G. (2006) Global analysis ofprotein palmitoylation in yeast. Cell 125, 1003–1013

49. Pica, N., and Palese, P. (2013) Toward a universal influenza virus vaccine:prospects and challenges. Annu. Rev. Med. 64, 189 –202

Differential Acylation of Influenza HA

DECEMBER 12, 2014 • VOLUME 289 • NUMBER 50 JOURNAL OF BIOLOGICAL CHEMISTRY 34989

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 13: Site-specific S-AcylationofInfluenzaVirusHemagglutinin · Hemagglutinin (HA) of influenza virus is a typical type I transmembrane glycoprotein with an N-terminal signal pep-tide,

Andrei V. Alexeevski and Michael VeitKatharina Brett, Larisa V. Kordyukova, Marina V. Serebryakova, Ramil R. Mintaev,

DECISIVE FACTOR FOR ATTACHMENT OF STEARATETHE ACYLATION SITE RELATIVE TO THE MEMBRANE BORDER IS THE

-Acylation of Influenza Virus Hemagglutinin: THE LOCATION OFSSite-specific

doi: 10.1074/jbc.M114.586180 originally published online October 27, 20142014, 289:34978-34989.J. Biol. Chem. 

  10.1074/jbc.M114.586180Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/289/50/34978.full.html#ref-list-1

This article cites 48 references, 19 of which can be accessed free at

by guest on Decem

ber 4, 2020http://w

ww

.jbc.org/D

ownloaded from