ebook img

Multiple domains of EBER 1, an Epstein-Barr virus noncoding RNA, recruit human ribosomal ... PDF

12 Pages·2006·0.78 MB·English
by  
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview Multiple domains of EBER 1, an Epstein-Barr virus noncoding RNA, recruit human ribosomal ...

JOBNAME:RNA12#52006PAGE:1OUTPUT:MondayApril315:40:152006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press Multiple domains of EBER 1, an Epstein-Barr virus noncoding RNA, recruit human ribosomal protein L22 VICTOR FOK,1,2,4 RACHEL M. MITTON-FRY,1,4 ANGIE GRECH,3 and JOAN A. STEITZ1,2 1DepartmentofMolecularBiophysicsandBiochemistryand2HowardHughesMedicalInstitute,YaleUniversitySchoolofMedicine, NewHaven,Connecticut06536,USA 3McKinseyandCompany,Inc.,NewYork,NewYork10022,USA ABSTRACT EBER1,asmallnoncodingviralRNAabundantlyexpressedinallcellstransformedbyEpstein-Barrvirus(EBV),hasbeenshown to associate with the human ribosomal protein L22. Here we present in vitro binding studies using purified RNAs and recombinant proteins. Electrophoretic mobility-shift assays (EMSAs) show that recombinant L22 (rL22) and maltose-binding protein (MBP)-tagged L22 protein bind EBER 1 in vitro, both forming three specific protein-dependent mobility shifts. Use of amixtureofrL22andMBP-L22indicatesthatthesethreeshiftscontainone,two,orthreeL22proteinsperEBER1molecule. EMSAs performed withEBER 1deletionconstructs and EBER1stem–loops insertedinto a nonbinding RNA,HSUR3, identify stem–loopsI,III,andIVasL22bindingsites.TheexistenceofmultipleL22bindingsitesonEBER1insidecellsisdemonstrated byin vivo UVcross-linking.Our resultsare discussedwith respect tothe function ofEBER1in EBV-infectedhuman Bcells. Keywords: Epstein-Barr virus; EBER 1; ribosomal protein L22; electrophoretic mobility-shift assay; in vivo UV cross-linking; RNA–protein interaction INTRODUCTION genesdeletedwasshowntoexhibit100-foldlowerefficiency in malignant transformation of resting B cells compared Epstein-Barr virus (EBV) is a member of the human withitswild-typecounterpart(Yajimaetal.2005),suggest- gherpesvirusfamilythatinfects>90%ofadultsworldwide ing a role for EBERs in cellular transformation. This and maintains a lifelong presence. EBV is the causative confirmsearlierreportsthatexpressionofEBERsalonepro- agentofinfectiousmononucleosis(Henleetal.1968)andis motesthetumorogenicphenotypeasassessedbycellgrowth associated with Burkitt lymphoma (BL) (Henle and Henle insoftagarandtumorgenicityinnudemice(Komanoetal. 1966),nasopharyngealcarcinoma(zurHausenetal.1970), 1999;Rufetal.2000;Yamamotoetal.2000).However,the and other human tumors including Hodgkin’s disease molecular mechanisms underlying the function(s) of the (Johansson et al. 1970, 1975) and AIDS-associated lym- EBERs in the viral life cycle remain unclear. phoma (Subar et al. 1988; Haluska et al. 1989). While EBER 1 and EBER 2 (167 and 172 nt, respectively) have most of the EBV genome is silent in EBV-transformed considerablesecondarystructureconsistingofseveralstable B-lymphocytes, two genes encoding the small RNAs EBER stem–loops (Glickman et al. 1988). They contain 39 termi- 1 and EBER 2 (Epstein-Barr encoded RNAs) are actively nal polyuridylate stretches that interact stably with the transcribed by RNA polymerase III (z106 copies per cell) La antigen (Lerner et al. 1981). In addition to La, EBER 1 (Lerner et al. 1981; van Santen et al. 1981; Arrand et al. (Fig. 1A) binds the human ribosomal protein L22 tightly 1989). Recently, a recombinant EBV with the two EBER (Toczyski and Steitz 1991) and the interferon-inducible protein kinase R (PKR) more weakly (Clarke et al. 1990, 1991; Clemens et al. 1994). L22 is a 14.8-kDa protein con- 4Theseauthorscontributedequallytothiswork. stituent of the large ribosomal subunit, which is not Reprint requests to: Joan A. Steitz, Department of Molecular Bio- physics and Biochemistry and Howard Hughes Medical Institute, Yale conserved in prokaryotes (Nakao et al. 2004) and does University School of Medicine, New Haven, CT 06536, USA; e-mail: not share RNA binding motifs with any other known [email protected];fax:(203)624-8213. protein(http://us.expasy.org/prosite).Inaddition,L22asso- Articlepublishedonlineaheadofprint.Articleandpublicationdateareat http://www.rnajournal.org/cgi/doi/10.1261/rna.2339606. ciates with human telomerase (Le et al. 2000), the small 872 RNA(2006),12:872–882.PublishedbyColdSpringHarborLaboratoryPress.Copyright"2006RNASociety. A23396Foketal.ARTICLERA JOBNAME:RNA12#52006PAGE:2OUTPUT:MondayApril315:40:152006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press L22 bindsmultipledomains ofEBER1 50% of the L22 protein associates with this RNA (Toczyski et al. 1994). Pre- vious studies concluded that L22 binds EBER1viastem–loopIII(Toczyskiand Steitz1993;Fig.1A),butthatadditional binding site(s) may also exist (Toczyski and Steitz 1993; Dobbelstein and Shenk 1995). SELEX (systematic evolution of ligands by exponential enrichment) experiments established a generalized RNA motif (Fig. 1D) for L22 binding (Dobbelstein and Shenk 1995). Both stem–loops III and IV of EBER 1 fit this motif, thereby implicating stem– loopIVasanadditionalbindingsitefor L22 on EBER 1. To investigate the molecular interac- tions involved in RNP formation by EBER 1 and L22, we carried out more extensive binding studies using un- tagged and MBP-tagged L22 protein. EMSAs (electrophoretic mobility-shift assays) performed with wild-type EBER 1 and these recombinant proteins re- vealed three specific mobility shifts that FIGURE1. L22interactionswithEBER1andHSUR3inEMSAs.(A)Secondarystructureof aredependentonproteinconcentration wild-type EBER 1 as determined previously (Glickman et al. 1988). Bold roman numerals and differ in the ratios of protein/RNA denotethestem–loops.(B)SecondarystructureofHSUR3fromHerpesvirussaimiri(Leeetal. 1988). The arrow indicates where stem–loops I,II, III, or IVwere inserted into the RNA to in the shifted complexes. Examination generatetheHSUR3chimerasanalyzedinFigure5.(C)EMSAsperformedwith5nMbody- of EBER 1 deletion constructs and labeled HSUR 3 (lanes 1–6) or EBER 1 (lanes 7–11) and increasing concentrations of chimericRNAmoleculescontainingiso- recombinantL22protein.EBER1,whichalonemigratesasaclosedoublet,formsthreedistinct shiftswithL22,whileHSUR3shiftsonlyatproteinconcentrations$3mM.Anasterisk(*) latedEBER1stem–loopsinEMSAsiden- indicates an additional, nonspecific shift that appears at very high L22 concentrations. Note tified three L22 binding sites. These thatthefreeHSUR3containsaslower-migratingbandevenwithoutL22.(D)Thegeneralized include the above-mentioned stem–loops RNAconsensusmotifproposedtomediateinteractionbetweenL22andRNA(adaptedfrom IIIandIV,andapreviouslyundescribed DobbelsteinandShenk1995). site involving stem–loop I. In addition, we demonstrate the existence of multi- viral RNA HVP-1 in Herpesvirus papio–infected baboon pleL22bindingsitesonEBER1insidecellsbyaUVcross- cells (Toczyski and Steitz 1991), the 39X region within the linkingassay.UnderstandinghowL22associateswithEBER 39-untranslated region (UTR) of the human Hepatitis C 1 may help elucidate the contribution of the EBER RNPs virus(HCV)genomicRNA(Woodetal.2001),poly(ADP- to the EBV life cycle. ribose)polymeraseinDrosophilamelanogaster(Koyamaetal. 1999),andtheHerpessimplex–infectedcellproteins(ICP)4 RESULTS and ICP 22 in human cells (Leopardi and Roizman 1996; Leopardietal.1997).However,thefunctionofL22bothin Stem–loops I, III, and IV are all binding sites for rL22 the ribosome and in these several RNP complexes remains unknown. EMSAwas usedtostudytheinvitro interaction ofhuman In uninfected human B cells, L22 is located in the ribosomal protein L22 with EBER 1 in comparison to an nucleoli and cytoplasm, whereas following EBV infection, unrelated RNA derived from Herpesvirus saimiri, HSUR 3 L22 is found in the nucleoplasm as well (Toczyski et al. (Lee et al. 1988). Increasing concentrations of untagged 1994).InsituhybridizationindicatesthatEBERsarelargely recombinant L22 (rL22) were incubated with either 5 nM nucleoplasmic(HoweandSteitz1986;Barlettaetal.1993), 32P-labeled full-length EBER 1 (Fig. 1A) or HSUR 3 (Fig. suggesting that L22 relocalization results from its associa- 1B). Three distinct protein-dependent mobility shifts were tion with EBER 1 in vivo (Toczyski et al. 1994). In trans- observed for EBER 1 (Fig. 1C, lanes 8–11), suggesting that formed B cells, EBER 1 is quantitatively bound by L22 the dominant species present at 1 mM rL22 is the triply (>95%) (Toczyski and Steitz 1993), whereas only 30%– boundRNA. Anadditional diffuseshift canalsobeseenat www.rnajournal.org 873 JOBNAME:RNA12#52006PAGE:3OUTPUT:MondayApril315:40:172006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press Fok etal. 3 mM rL22. In contrast, HSUR 3 begins to shift at protein previously indicated that stem–loop III of EBER 1 binds concentrationsonlyatorabovethislevel(Fig.1C,lanes2– L22 (Toczyski and Steitz 1993), and SELEX and EMSAs 6). Collectively, these data argue that EBER 1 shifts 1, 2, (Dobbelstein and Shenk 1995) suggested stem–loop IV as and3(Fig.1C,lanes8–11)representspecificrL22–EBER1 an additional L22 protein binding site. To verify these and complexes, whereas the additional diffuse shift most likely identify other RNA elements within EBER 1 important for arises from nonspecific electrostatic association with the binding L22, we performed a series of mobility-shift ex- RNA at high protein concentrations. periments using EBER 1 deletion constructs in which each The three distinct mobility shifts (Fig. 1C) suggest that of the stem–loops was deleted independently (Fig. 2B–E). rL22 forms at least three distinct complexes withwild-type Interestingly, single deletion of three of the stem–loops EBER1.Oneinterpretationisthatwild-typeEBER1possesses of EBER 1 pictured in Figure 1A abolished or greatly three unique L22 binding sites. However, it cannot be hindered the formation of the three specific shifts. These assumed that each shift represents a unique molecular were stem–loop I (EBER 1 Dstlp I, Dnts 14–33; Fig. 2B), complex;asingleshiftcouldcontainheterogeneousmixtures stem–loopIII(EBER1DstlpIII,Dnts60–83;Fig.2D),and ofdifferentcomplexesthattravelwiththesamemobilityin stem–loop IV (EBER 1 Dstlp IV, Dnts 87–123; Fig. 2E). In native polyacrylamide gels. Immunoprecipitation studies contrast, deletion of stem–loop II from EBER 1 (EBER 1 FIGURE2. EMSAsperformedwithEBER1deletionconstructsidentifystem–loopsI,III,andIVasbindingsitesforL22onEBER1.Predicted secondary structures of the EBER 1 deletion RNAs tested (top) and EMSAs performed with 5 nM RNA (body-labeled) and increasing concentrationsofL22(bottom).(A)Full-lengthEBER1,(B)EBER1DstlpI(Dnts14–33),(C)EBER1DstlpII(Dnts45–58),(D)EBER1DstlpIII (Dnts60–83),(E)EBER1DstlpIV(Dnts87–123).Thedeletionmutantsarenumberedaccordingtothefull-lengthRNA.Anasterisk(*)indicates anadditional,nonspecificshiftthatonlyappearsatL22concentrations$3mM. 874 RNA, Vol.12, No.5 JOBNAME:RNA12#52006PAGE:4OUTPUT:MondayApril315:40:212006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press L22 bindsmultipledomains ofEBER1 DstlpII,Dnts45–58;Fig.2C)producedanRNAwithrL22- presence of either rL22 or MBP-L22 alone, three specific bindingbehaviorqualitativelyidenticaltothatofwild-type concentration-dependentbandsappeared(Fig.3,lanes2–5 EBER1(Fig.2A).Thesedifferencesarebestexemplifiedby or9–11,respectively).WhenEBER1DstlpIIwasincubated examinationofEBER1shiftsinthepresenceof1mMrL22. withamixtureofthetwoproteins(Fig.3,lanes6–8),these Themajorityofthesignalfromfull-length(Fig.2A,lane4) individual shifts were observed, as well as three distinct and Dstlp II RNAs (Fig. 2C, lane 5) is found in shift 3. In intermediate shifts. The formation of a band intermediate contrast, the bulk of the signal from EBER 1 Dstlp I (Fig. to a shift seen with each protein alone would suggest that 2B,lane4)orEBER1DstlpIII(Fig.2D,lane5)isfoundin two (or more) molecules of protein bind. shift1,whilethatfromEBER1DstlpIV(Fig.2E,lane5)is The simplest interpretation of the above mixing exper- found in shift 2. iment(Fig.3)isthatproteinshifts1,2,and3containone, These results corroborate the previous findings that two, and three of the relevant L22 molecules bound to stem–loops III and IV are essential for full rL22 binding EBER 1, respectively, while the intermediate shifts contain toEBER1andsuggestanadditional,previouslyundescribed both rL22 and MBP-L22 molecules simultaneously bound protein binding site in stem–loop I. It was surprising, to EBER 1. Mixed shift 1 represents EBER 1 bound to two however, that deletion of stem–loop III did not appear to L22 moieties (one molecule of rL22 and one molecule of retard the formation of the first protein shift. In fact, the MBP-L22), while mixed shifts 2 and 3 represent EBER 1 first protein shift was observed even at the lowest concen- bound to three L22 moieties (two rL22 and one MBP-L22 tration(0.1mM)shownforallsingle-loopdeletionconstructs, moleculesinmixedshift2,andonerL22andtwoMBP-L22 suggesting malleability in the rL22–EBER 1 interaction. molecules in mixed shift 3). Although both mixed shifts 2and3containthreeproteinmolecules,thelattermigrates significantly more slowly, consistent with its proposed EBER 1 binds three protein molecules simultaneously greater bulk. The protein-concentration dependence of the appearance of the mobility shifts, in concert with the presence of EBER 1 fragments bind rL22 binding sites on stem–loops I, III, and IV, suggests that EBER 1 can bind multiple copies of rL22 simultaneously. To probe the rL22-EBER 1 interactions further, we per- Todeterminetheprotein/RNAstoichiometryforshifts1–3, formed mobility-shift assays with several EBER 1 frag- binding studies were performed using a mixture of rL22 ments. EBER 1 nts 45–124, which comprises stem–loops (15 kDa) and MBP-L22 fusion protein (56 kDa) (Fig. 3). II–IV of EBER 1, formed two shifts with rL22. These shifts EBER 1 Dstlp II was substituted for full-length EBER 1, as appeared at similar rL22 concentrations to the first two it exhibited very similar binding behavior with both rL22 shifts of full-length EBER 1. Shortening stem–loop IV (Fig. 2A,C) and MBP-L22 (data not shown). However, the reducedtheassociationof theRNA fragmentwiththefirst increasedresolutionwiththissmallerRNAbetweenbound protein molecule and abrogated the second shift (data not speciesgreatlyfacilitatedinterpretationoftheresults.Inthe shown),supportingtheconclusionthatstem–loopIVisan rL22-binding site. Identical results were seen upon sub- stituting theloopofstem–loop II, whichwasshown above not to be essential for protein binding, with a GAAA tetraloop(datanotshown).Ourrationalewasthatasmaller construct comprised of just stem–loops II and III would usethisGAAAtetraloopinstem–loopIItopromoteproper RNA folding. Again, this construct showed only one shift with rL22 (Fig. 4B), but with slightly reduced affinity for rL22. This suggests that elements in the base of stem–loop IV might be necessary either for direct binding to rL22 or for correct presentation of stem–loop III to the rL22- binding surface. Similar results were seen upon removal of stem–loop II entirely (Fig. 4C). FIGURE 3. Shifts 2 and 3 contain multiple L22 molecules. EMSAs AnotherseriesofEBER1fragmentsremovedstem–loops performedwith5nMbody-labeledEBER1DstlpIIandtheindicated I, II, and IV from the backbone of the RNA molecule. concentrationsofrL22and/orMBP-L22fusionprotein.EBER1Dstlp Unexpectedly, EBER I Dstlps I, II, and IV formed two II was chosen for the mixing experiment, as it exhibits L22-binding behaviorsimilartofull-lengthEBER1butbetterseparationbetween RNA–protein complexes in mobility-shift assays (Fig. 4D). protein-bound species. Mixed shift 1 is presumed to comprise one This behavior may be explained by the presence of two rL22andoneMBP-L22moleculeboundtotheRNA,withtworL22 energetically comparable RNA conformers, one of which andoneMBP-L22moleculesinmixedshift2,andonerL22andtwo containsanovelbindingsiteforL22(seeDiscussion).Short- MBP-L22moleculesinmixedshift3.Anasterisk(*)indicatesaminor multiplyshiftedspecies. ening the terminal RNA stem abolished the second shift www.rnajournal.org 875 JOBNAME:RNA12#52006PAGE:5OUTPUT:MondayApril315:40:222006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press Fok etal. FIGURE 4. EMSAs investigating the interaction of L22 with EBER 1 fragments. Predicted secondary structures of EBER 1 truncations and EMSAsperformedwith5nMRNAandincreasingconcentrationsofL22areshown.(A)EBER1nts45–124,(B)EBER1nts45–84,GAAA,(C) EBER1nts59–87,(D)EBER1DstlpsI,II,andIV,and(E)EBER135–135DstlpsIIandIV.Thedeletionmutantsarenumberedaccordingtothe full-lengthRNA.EMSAswereperformedwithbody-labeledRNAs,withtheexceptionoftheconstructsinAandE,whichwere59-endlabeled.An asterisk(*)indicatesanadditional,nonspecificshiftthatonlyappearsatveryhighL22concentrations.AsstlpIIwasshownnottobeinvolvedin L22binding,itsloopwasreplacedwithaGAAAtetralooptopromotecorrectfoldingoftheconstructstudiedinB. without affecting the binding affinity of the first shift (Fig. HSUR3eachproducedasingle-shiftedcomplexwithrL22. 4E; data not shown). The same amounts (1 and 4 mM) of rL22 did not yield aspecificshiftwithHSUR3alone(Fig.5,lanes3,4)orwith Stem–loops I, II, and IV can individually bind rL22 the HSUR 3 stem–loop II chimera (data not shown). We AlthoughthedatainFigures2and4arguethatstem–loops concludethatstem–loopsIandIVcanindependentlyserve I,III,andIVarenecessaryforL22bindingtoEBER1,they as binding sites for rL22. Interestingly, the HSUR 3 stem– donotaddresswhethertheseregionsaresufficient.Because loop III (nts 59–84) chimera consistently yielded two stem–loop III alone binds poorly to rL22 even at 10 mM differently migrating forms of the free RNA (Fig. 5, lane 8) concentration (data not shown) and decreases in binding inmultipleRNApreparationspurifiedfrompolyacrylamide affinitywereobservedwhenstem–loopIVwasdeleted(Fig. gels, arguing that it adopts two major conformations (see 4,cf.AandB/C),wereasonedthatthestem–loopsofEBER Discussion). Increasingamounts ofrL22shiftedbothRNA 1 might require the context of a larger RNA molecule to conformersintospecificcomplexes(Fig.5,lanes9–11)but fold properly. We therefore performed binding assays on with more background than the profiles of the HSUR 3 the individual EBER 1 stem–loops inserted (see Fig. 1B for stem–loop I or IV chimeras. The diminution of the free siteofinsertion)intothenonbindingHSUR3moleculeto HSUR 3 stem–loop III band even at 0.4 mM rL22 (Fig. 5, form HSUR 3 stem–loop chimeras. lanes 9–11) argues that stem–loop III is an L22-binding TheinsertionofEBER1stem–loopI(nts14–33)(Fig.5, site.Takentogether,thesedatasupporttheideathatEBER lanes 6,7) or IV (nts 87–123) (Fig. 5, lanes 14,15) into 1 stem–loops I, III, and IV can individually bind L22. 876 RNA, Vol.12, No.5 JOBNAME:RNA12#52006PAGE:6OUTPUT:MondayApril315:40:252006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press L22 bindsmultipledomains ofEBER1 DISCUSSION In vitro binding experiments presented here demonstrate that wild-type EBER 1 forms three distinct mobility shifts with rL22 or MPB-L22 that are protein-concentration dependent (Fig. 1; data not shown). The association is direct since it occurs in the absence of any other viral or host proteins and does not require post-translational modifications. The three distinct RNP complexes observed differinthetotalnumberofboundL22proteinmolecules; shifts1,2,and3appeartocontainone,two,andthreeL22 molecules bound to EBER 1, respectively (Fig. 3). Because thedeletionofEBER1stem–loopI,III,orIVgreatlyaffects theformationofathirdcomplex(Fig.2),thesethreestem– FIGURE 5. L22 interacts with HSUR 3 chimeras containing in- loopsappeartocomprisethethreebindingsitesforL22in dividual EBER 1 stem–loop sequences. EMSAs were performed with vitro. Indeed, our data agree with previous observations 5 nM HSUR 3 without an insert (lanes 1–3) and HSUR 3 chimeras containingeitherstem–loopI(lanes4–7),stem–loopIII(lanes8–11), that stem–loops III and IV are L22 binding sites (Toczyski or stem–loop IV (lanes 12–15) in the presence of the indicated andSteitz1993;DobbelsteinandShenk1995).Insertionof concentrations of rL22 protein. Note that HSUR 3 stem–loop III stem–loop I into HSUR 3 confers binding activity on this containstwoformsofthefreeRNA;theirrespectiveshiftsaremarked unrelatednonbindingRNA,confirmingthatstem–loopIis withsingle(*)anddouble(**)asterisks.The**shiftsometimes,but notalways,appearsasadoublet. abindingsiteforL22(Fig.5).Finally,invivocross-linking experiments show that there are also multiple L22-binding sites on EBER 1 inside cells (Fig. 6). EBER 1 binds multiple L22 Proteins in vivo HavingestablishedthatEBER1containsthreeL22-binding Can stem–loops be assigned to particular shifts? sitesinvitro,wewishedtodeterminewhethermultipleL22 proteins likewise interact with EBER 1 inside transformed With the wild-type EBER 1 molecule, we estimate the cells where other EBER 1-binding proteins could poten- apparent binding constants (K ) for the first, second, and d tiallycompete.WeusedapowerfulinvivoUVcross-linking third shifts to be 0.1, 0.3, and 1 mM, respectively. In an method (Cook et al. 2004) in which anti-L22 antibody was attempttoassigntheseshiftstoparticularbindingsites,we used to immunoprecipitate cross-linked EBER 1–L22 com- first examined fragments of EBER 1. We constructed two plexes under semidenaturing conditions. When a DNA small stem–loop III-containing fragments, EBER 1 nts 45– construct expressing wild-type EBER 1 and EBER 2 from 84, GAAA (Fig. 4B) and EBER 1 nts 59–87 (Fig. 4C), and their native promoters was transfected intoHEK293cells,EBER1wasdetected in the anti-L22 precipitate in a cross- linking-dependent manner (Fig. 6, top, cf. lanes 8,18 and 7,17). IfEBER1bindsmultiplecopiesofthe L22 protein in vivo, deletion of any one binding site from the RNA should not abolishprecipitationafterUVcross-link- ing. Indeed, upon expression in HEK 293 cells and UV cross-linking, EBER 1 Dstlp I (Fig. 6, top, lane 10), EBER 1 FIGURE6. InvivointeractionsbetweenL22andEBER1deletionconstructs.HEK293cells Dstlp III (Fig. 6, top, lane 12), and weretransfectedwiththefollowingEBER1andEBER2expressionconstructs:wild-typeEBER EBER 1 Dstlp IV (Fig. 6, top, lane 20) 1 (lanes 1,2,7,8,13,14,17,18), Dstlp I (lanes 3,4,9,10), Dstlp III (lanes 5,6,11,12), or Dstlp IV weredetectedintheanti-L22precipitate. (lanes15,16,19,20).Intactcellswereirradiatedwith254-nmUVlighttoinducecross-linking The specificity of these interactions is between protein and RNA in vivo (even-numbered lanes); untreated cells served as negative controls (odd-numbered lanes). Whole-cell lysates were subjected to immunoprecipitation demonstrated by the absence of EBER 2 with anti-L22 antibody under denaturing conditions. Precipitated RNAs were released from in the precipitates (Fig. 6, bottom). We theantibodybyproteinaseKdigestionandwereresolvedinan8%ureapolyacrylamidegel.A concludethatmultipleL22moleculesare totalof0.5%oftheinputand100%oftheprecipitateswereanalyzed,andEBER1sequences weredetectedbyNorthernblotting(top).ThelowexpressionlevelsofEBER1DstlpI(lanes boundtoEBER1insidecells,asinvitro, 3,4)andDstlpIII(lanes5,6)areduetothedisruptionofBoxAorBoxBoftheEBER1RNA but we cannot distinguish whether two polymeraseIIIpromoter,respectively.Tocontrolfornonspecificcross-linkingandimmuno- or more L22-binding sites exist. precipitation,themembraneswerestrippedandreprobedforEBER2sequences(bottom). www.rnajournal.org 877 JOBNAME:RNA12#52006PAGE:7OUTPUT:MondayApril315:40:272006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press Fok etal. estimatedthatstem–loopIIIhasaK of1mM.However,in tionofanystem–loopfromtherestofthemolecule(Fig.5) d amoleculethatcontainsstem–loopsII,III,andIV(EBER1 negatively affects the interaction between L22 and EBER 1 nts 45–124;Fig. 4A) the first and second shifts haveK sof stem–loops. Therefore, we conclude from our results only d 0.1 and 1 mM, respectively. These affinities suggest that that wild-type EBER 1 has three in vitro L22-binding sites, shift 1 with EBER 1 nts 45–124 corresponds primarily to whoserelativeaffinities remain unknown. It is quite possi- L22 binding at stem–loop IV, with an additional L22 ble that each shift represents a mixture with L22 bound to molecule at stem–loop III to form shift 2. Alternatively, several of its binding sites. additionalsequencesfromstem–loopIVmightallowstem– loop III to bind L22 with a higher affinity, perhaps by What molecular features does L22 recognize? facilitating proper RNA folding. In this case, shift 1 on SELEX experiments predicted a consensus RNA structure EBER 1 nts 45–124 would correspond primarily to L22 that enables specific interactions between L22 and RNA binding at stem–loop III. (Dobbelstein and Shenk 1995; Fig. 1D). The domains of We used M-fold (Zucker 2003) to predict RNA second- EBER 1 that fit this motif best are stem–loops III and IV. ary structures for wild-type EBER 1 and all of the deletion Sincestem–loopIIIwasalreadyestablishedasabindingsite constructRNAsusedinFigures2and4.M-foldconfirmed (Toczyski and Steitz 1993), Dobbelstein and Shenk (1995) thestructuresshowninallcasesexceptforEBER1DstlpsI, proposed stem–loop IV as the additional binding site on II, and IV (Fig. 4D). Here, an alternate structure of EBER 1. EMSAs performed with isolated stem–loop IV in comparable predicted stability can form by refolding nts theabsenceofcompetitorRNAindeeddemonstratedweak 127–138tocreateahairpinloopwitha4-bpstemclosedby bindingthatcouldbecompetedbytheadditionofisolated GC and a U in the 39 position of the loop, conforming to stem–loop III (Dobbelstein and Shenk 1995). theconsensusmotifforL22binding(Fig.1D).L22binding Herewehaveidentifiedstem–loopIofEBER1asathird both to stem–loop III and to this novel stem–loop in the binding site for L22. This assignment is supported by molecule would then explain the two shifts seen in the a comparison of the EBER 1 stem–loop I to a sequence EMSA profile of this molecule (Fig. 4D). Moreover, this from the 39X sense strand of HCV genomic RNA that is structure would not form with EBER 1 35–135 Dstlps II reported to bind L22 (Wood et al. 2001); nt 19–25 within and IV (Fig. 4E), explaining loss of the second shift. the 39X sense strand are identical to nt 13–19 in EBER 1 Because individual stem–loops of EBER 1 may require stem–loopI(Woodetal.2001).Yet,stem–loopIofEBER1 the context of a larger RNA molecule to fold properly, we doesnotfitthepredictedconsensussequence(Dobbelstein also inserted them into a nonspecific RNA, HSUR 3. andShenk1995)entirely(seeFig.1A,D);itmaintainsaGC Curiously,theHSUR3stlpIIIchimeraassumedtwomajor base pairat theapicalend,withtheG59andtheC39,but conformations (Fig. 5, lane 8), again predicted by M-fold lacks the U residue in the 39 position of the loop. A (data not shown). While the two folds are dramatically commonfeatureisthatthestemsofbothstem–loopsIand different, they both retain stem–loop III. Thus, both con- III consist of 6 canonical bp, with no bulged residues. The formers are shifted (Fig. 5, lanes 9–11) and the estimated identification of more recognition sites for L22, including K s, based on the reduction of the free RNA signals, are d that in the large subunit of the ribosome, would provide <1 mM. The estimated K for the HSUR 3 stlp IV chimera d additional insights into the consensus-binding sequence (whose predicted structure is consistent with M-fold) is and structure. 2mM(Fig.5).However,forHSUR3stlpI,M-foldpredicts fourisoenergeticstructures,threeofwhichlackstem–loopI. Significance of the association between EBER 1 Its apparent Kd of >4 mM (Fig. 5, lanes 4–7) is therefore and ribosomal protein L22 not meaningful. Thus, we can only conclude that, when inserted into HSUR 3, stem–loop III exhibits stronger L22 ThehighabundanceofEBER1andEBER2inEBV-infected bindingthanstem–loopIV.Itisstrikingthattheestimated cells,togetherwiththeirevolutionaryconservation(Arrand K sfortheindividualstem–loopchimeras(Fig.5)areallat and Rymo 1982; Howe and Shu 1988), strongly suggests d least fourfold lower than when the same stem–loops are that these RNAs contribute importantly to the EBV life part of EBER 1 (Figs. 1C, 2A). cycle.TheassociationbetweenEBER1andribosomalprotein Given the results with both the EBER 1 fragments and L22 immediately suggests that the EBERs might modulate with the HSUR 3-EBER 1 chimeras, can we assign the translation. Based on the estimate that the intracellular various shifts to L22 interactions with specific stem–loops? concentration ofL22(z50mM)is10-foldthat ofEBER1, Figure2showsthattheindividualdeletionofstem–loopsI, nearly all of EBER 1 is predicted to be bound by multiple III, or IV surprisingly has no effect on the appearance of molecules of L22 in vivo. This is confirmed by the in vivo shift1.Thus,itisimpossibletodeducewhichstem–loopis cross-linkingdatainFigure6.SincedepletionofL22fromthe thestrongestbindingsiteforL22inthecontextofthefull- ribosomesofEBV-transformedcellswasnotexperimentally length RNA. Together our results indicate that any major observed (Toczyski et al. 1994), L22 levels may be selec- (Fig. 4) or minor (Fig. 2) deletion of EBER 1 or the isola- tively up-regulated in EBV-infected cells. 878 RNA, Vol.12, No.5 JOBNAME:RNA12#52006PAGE:8OUTPUT:MondayApril315:40:272006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press L22 bindsmultipledomains ofEBER1 ItisalsopossiblethatthesequesteringofL22byEBER1 above.EBER135–135DstlpII,IVwasgeneratedbyPCRampli- is not important, but rather that the RNP consisting of fication using EBER 1 Dstlps (I, II, and IV) plasmid as the DNA EBER1,L22,andadditionalhostproteinsfulfillssomespe- template and the following primers: 59 primer-GGGAATTCTAA cificfunctioninEBV-transformedcells.EBER1hasbeenshown TACGACTCACTATAGGAGACGUGUGUGGCUGU, whichcon- tains anEcoRI restriction sitefollowed byaT7RNApolymerase tointeractstablywiththeLaantigen(Lerneretal.1981)and promoter,and39primer-CGCCGGATCCTTTAAAGACGGCAGA PKR (Clarke et al. 1991; Sharp et al. 1993; Clemens et al. CACCACC,whichcontainsDraIandBamHIrestrictionsites.The 1994;Eliaetal.1996;ClemensandElia1997;Vuyisichetal. resulting product was cloned into a pUC19 plasmid using the 2002).Laissufficientlyabundant(z23107molecules/cell) EcoRIandBamHIrestrictionsites.EBER145–124wasgenerated that binding by EBERs (z106 molecules/cell) should not by PCR amplification using wild-type EBER 1 plasmid and the sequester La away from other roles in the cell (Wolin and followingprimers:59primer-GGGAATTCTAATACGACTCACTA Cedervall 2002). On the other hand, PKR is much less TAGUGGCUGUAGCCACCC, which contains an EcoRI restric- abundant(ClemensandElia1997)anditsbindingbyEBER tion site, followed by a T7 RNA polymerase promoter, and 39 1 could block its ability to inhibit protein synthesis in primer-CGCCAAGCTTTAAAGCAGAGUCUGGGAAGAC,which infectedcells (Clarke et al. 1991; Sharp etal. 1993; Clemens containsDraIandHindIIIrestrictionsites.Theresultantproduct et al. 1994).Recently,this model has beenextended to sug- was cloned into a pUC19 plasmid using the EcoRI and HindIII restriction sites. For EBER 1 nts 45–84, GAAA and EBER 1 nts gest that L22 may exert a protective effect against the 59–87,DNAtemplatescontainingaT7RNApolymerasepromoter transforming potential of EBER 1 in vivo by making EBER followed by the relevant sequence were synthesized by the Keck 1 unavailable for binding to PKR (Elia et al. 2004). This Facility at Yale University and purified on a 15% denaturing hypothesis rests on the idea that L22 and PKR compete for polyacrylamidegel.Stem–loopswereaddedtoHSUR3byligating a common binding site on EBER 1, namely stem–loop IV twoDNAinserts(59and39fragments)intotheHSUR3construct (Dobbelstein and Shenk 1995; Vuyisich et al. 2002), while usingtheEcoRIandSmaIrestrictionsites.TheHerpesvirussaimiri otherstudies,incontrast(Rufetal.2005;Wangetal.2005), HSUR 3 plasmid was previously constructed (Myer et al. 1992) do not find evidence for interactions between EBER 1 and andwasusedasPCRtemplate.The59fragmentsofHSUR3stlpI, PKR. Clearly, there is much more to be learned concerning HSUR 3 stlp III, and HSUR 3 stlp IV were generated by PCR the role of the EBER 1 RNP in EBV-transformed cells. amplificationwith59-CCTCTAGGGCTTGGGTTGTTAATCTCCTA, 59-TGTACCCGGGACGGCTTGGGTTGTTAATCTCCTA, and 59-CAACCACAGACACCGTCCTCCTTGGGTTGTTGTTAATCTCCTA, respectively, and T7 primer. The 39 fragments of HSUR 3 stlp I, MATERIALS AND METHODS HSUR 3 stlp III, and HSUR 3 stlp IV were generated by PCR amplification with 59-TTTTGCTAGGGAATAATTTTTGAAGGC Plasmid construction TCTGG,59-AGTCCCGGGTGGAATAATTTTTGAAGGCTCTGG, and 59-TCTTCCCAGACTCTGCTTTAATAATTTTTGAAGGCTC Wild-type EBER 1 was previously cloned into a pUC119 vector TGG, respectively, andSP6 primer. (Toczyski and Steitz 1993). This gene fragment (225 nt) was For the in vivo UV cross-linking experiments, EBER 1 and its removed from the pUC119 vector and placed into a pUC19 deletionvariantswereexpressedusingtheirendogenouspromoters vector. Briefly, the EBER 1 gene (167 nt), preceded by an EcoRI from a plasmid encoding both EBER 1 and EBER 2. p73EBERs restriction site and a T7 RNA polymerase promoter (23 nt) and was constructed by ligating a 1.1-kb DNA fragment from the followed by a HindIII restriction enzyme site, was excised from EcoRIJfragmentofEBVintopSP73(Promega)usingtheSacIand the pUC119 vector by cleavage with the enzymes EcoRI and BamHIrestrictionsites.Stem–loopsweredeletedfromEBER1by HindIIIandinsertedintotheEcoRIandHindIIIsitesofpUC19. ligating two DNA inserts (59 and 39 fragments) into p73EBERs All EBER 1 stem–loop deletion plasmids were synthesized using usingtheEcoRIandBamHIrestrictionsites.The59fragmentsof the QuikChange Site-Directed mutagenesis kit (Stratagene) Dstlp I, Dstlp III, and Dstlp IV were generated by PCR amplifi- according to the manufacturer’s protocol. Wild-type EBER 1 cation with 59-AGGAGACGTGTGTGGCTG, 59-TGAGGACG plasmid was used as the DNA template in all cases, unless GTGTCTGTGG,and59-TCTGCCGTCTTCGGTCAA,respectively, otherwise specified. The following 59 oligonucleotides were used and an SP6 primer. The 39 fragments of Dstlp I, Dstlp III, and tocreatethecorrespondingdeletionconstructs(with39oligonu- Dstlp IV were generated by PCR amplification with 59-GCG cleotides complementary to their respective 59 oligonucleotides): TAGGTCCTGAGGTTCA, 59-GTGGCTACAGCCACACAC, and EBER 1 Dstlp I, 59-AGGACCTACGCTGAGGAGACGTGTGTGG 59-CACCACCCGGGACTTGTACC,respectively,andaT7primer. CTG;EBER1DstlpII,59-GCTAGGGAGGAGACGTGTCCGTCC DNAsequences ofallplasmidswereverifiedbytheKeckFacility CGGGTACAAGT;EBER1DstlpIII,59-CGTGTGTGGCTGTAGC at YaleUniversity. CACCGTGAGGACGGTGTCTGTG.EBER1DstlpIVwasconstructed in two rounds. The first round removed nt 106–123 using 59-GGACGGTGTCTGTGGTTGTCTGCCGTCTTCGGTC.Thesec- Protein expression and purification ond round used this EBER 1 plasmid as the DNA template and removed nt 87–105 using 59-CAAGTCCCGGGTGGTGTCTGCC OverexpressionofrecombinantL22(rL22)andMBP-L22proteins GTCTTCGGTC.EBER1Dstlps(I,II,andIV)usedEBER1Dstlp followedasimilarprocedure. Atotalof0.4–1.2LofLBmedium IVplasmidastheDNAtemplateandremovedstem–loopsIandII containing 50 mg/mL ampicillin and 34 mg/mL chloramphenicol sequentially using the corresponding oligonucleotides described wereinoculatedwith0.5–1mLofanovernightcultureofRosetta www.rnajournal.org 879 JOBNAME:RNA12#52006PAGE:9OUTPUT:MondayApril315:40:272006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press Fok etal. (DE3)pLysScells(Novagen)harboringanL22expressionplasmid. linearized with DraI, which yields a 39 end identical to that of Cells were grown at 37°C for 3.5–5 h to an A of 0.6–0.7, and EBER1.HSUR3andHSUR3chimeraswerelinearizedwithSmaI 595 IPTG was added to a final concentration of 1 mM. Protein andPmlI,respectively,resultingintheadditionofaGnucleotide expression was allowed for 2 h at 37°C. Cells were harvested by at the 39 end. To obtain unlabeled RNAs, in vitro transcription centrifugation andresuspended atA of30–60in50mMTris- was performed overnight at 37°C in a final volume of 100 mL 595 HClatpH8.0/10%sucroseforrL22orincolumnbuffer(20mM containing 40 mM Tris-HCl (pH 7.5), 15 mM MgCl , 4 mM 2 Tris-HCl at pH 7.4, 200 mM NaCl, 1 mM EDTA, and 10% spermidine, 0.05% Triton X-100, 10 mM DTT, 2.5 mM NTPs, glycerol) for MBP-L22 proteins before being stored at "80°C. 0.05 mg/mL linearized plasmid or DNA template, and 0.1 mg/mL Frozen cells were thawed in an ice-water bath. All subsequent T7 RNA polymerase. The RNA transcripts were purified on 8% stepswerecarriedoutat4°C.Proteaseinhibitorcocktail(Calbio- denaturing polyacrylamide gels. Gel slices were eluted overnight chem) and a final concentration of 1–4 mM EDTA were added inTEbuffer(10mMTris-HClatpH7.5,1mMEDTA)containing to the thawed cells. Lysed cells were cleared by centrifugation at 1% SDS, and then phenol-extracted (pH 4.5), chloroform- 100,000g for 1 h at 4°C. Both L22 proteins were purified as extracted twice, and ethanol-precipitated in 0.3 M NaOAc (pH described below to 90%–95% homogeneity as judged by SDS- 4.5). Precipitated RNAs were washed with 70% ethanol and PAGE. The calculated extinction coefficients (e ) for rL22, dissolved in water. The RNA concentration was determined by 280 1209 M"1 cm"1, and for MBP-L22, 76,810 M"1cm"1, were used measuring theUV absorbanceat 260nm.A totalof50 rmolsof to determine proteinconcentrations. each transcript were treated for 1 h at 37°C with 0.5 U/mL calf intestinal alkaline phosphatase (Roche) to remove the 59 tri- phosphate. The RNA was extracted with phenol (pH 4.5), Purification of recombinant L22 protein followedbyachloroformextractionandanethanolprecipitation. It was resuspended in water and 59 phosphorylated by adding Plasmid pET11a-L22 constructed by PCR amplification of the 10 U of T4 polynucleotide kinase (PNK), 1 3 T4 PNK buffer L22-encoding sequence and ligation into the pET11a vector was (Roche), and 150 mCi of [g-32P]ATP for 45 min at 37°C. usedforexpressionofrL22.TheDNAsequencewasconfirmedby Radiolabeled transcripts were purified by separation on denatur- the Keck Facility. Cleared cell lysates were adjusted to Buffer A ingpolyacrylamidegels,elutionfromgelslices,andprecipitation (50 mM HEPES at pH 7.9, 4 mM EDTA, and 10% glycerol) asdescribed above. Thepelleted RNAswere dissolvedin water. containing 200mMNaClandloadedontoa1-mLSP-Sepharose To produce [a-32P]UTP body-labeled RNAs, in vitro tran- FastFlowcolumnequilibratedwiththesamebuffer.Thecolumn scription wasperformedfor 90minat37°Cinafinal volumeof was washed with 1 mL Buffer A containing 200 mM NaCl and 10 mL containing 40 mM Tris-HCl (pH 7.5), 6 mM MgCl , then 10 mL Buffer A containing 400 mM NaCl. rL22 was eluted 2 2 mM spermidine, 10 mM DTT, 0.5 mM ATP and CTP, 0.5 or in 6 mL Buffer A containing 900 mM NaCl. The purified rL22 1.0mMGTP,0.1or0.05mMUTP,20or40mCiof[a-32P]UTP protein was dialyzed twice against 25 mM HEPES (pH 7.9), (400 Ci/mmol), 0.1 mg/mL linearized plasmid or 6 ng/mL DNA 50 mMNaCl, 2 mMDTT,and 10%glycerol for 4–12h. template, and 0.1 mg/mL T7 RNA polymerase. RNAs were purified asdescribedabove. Cloning and purification of MBP-L22 fusion protein Electrophoretic mobility-shift assays TheL22encodingsequencewasPCRamplifiedfromtheGST-L22 plasmid (Toczyski and Steitz 1993) using a 59 primer-GCGGA The 59-end-labeled or [a-32P]UTP body-labeled RNAs (5 nM) ATTCGCCCCCGTGAAGAAGCTGTGanda39primer-TTTCAC wereheatedat95°Cfor3min,followedbysnapcoolingonicefor CGTCATCACCGAA. The resulting PCR product was restriction 3min.Bindingreactions(10mL)containingtheindicatedamounts digestedwithEcoRIandSalIenzymesandligatedintopMAL-c2X of protein were incubated for 30 min on ice in RNA-binding vector(NEB)cutwiththesameenzymes.TheMBP-L22sequence buffercontaining37.5mMHEPES(pH7.9),75mMNaCl,5mM wasconfirmed byDNAsequencing (Keck Facility). MgCl , 8% glycerol, and 1 mg Escherichia coli tRNA. Prior to 2 Before centrifugation, thawed cells were sonicated on ice for loading, 2 mL of 50% glycerol were added to each sample. The 10sectwice,20-secapart,withaBransonSonifieratsetting3and sampleswereloadedonapre-electrophoresed(200V,30min),8% 100% duty cycle. The soluble lysates were diluted with 20 mL nativegelin1XTBEbuffer(100mMTris,100mMborate,2mM column buffer and then loaded onto a 4-mL amylose column EDTA).Electrophoresiswascarriedoutat200Vfor3–6hat4°C. (NEB)equilibratedwithcolumnbuffer.Thecolumnwaswashed with60mLcolumnbufferandtheboundMBP-L22waselutedin UV cross-linking 6 mL column buffer containing 10 mM maltose. The purified MBP-L22proteinwasdialyzedtwiceagainst25mMHEPES(pH To detect in vivo cross-linking between L22 and RNA, 20 mg of 7.5), 50 mM NaCl, 2 mM DTT, and 10% glycerol for 4–8 h. A p73EBERs or its deletion variants were transfected into HEK293 fraction of the MBP-L22 preparation may have been inactive, cellsat40%–60%confluencyina10cm-diameterplateusingthe since rL22 exhibits about threefold higher binding affinity to TransIT293reagent(Mirus)accordingtothemanufacturer’spro- EBER1 ascompared withMBP-L22. tocol.After48h,thecellswerewashedtwicewithcoldphosphate- buffered saline and resuspended in 3 mL of phosphate-buffered In vitro transcription saline. Cells were irradiated (or not irradiated as a negative control) on ice with 254-nm UV light at a distance of 4 cm for RNAs for binding studies were prepared by in vitro run-off 4.5minandcollectedbyscrapingandcentrifugationinEppendorf transcription. Wild-type or deletion EBER 1 plasmids were tubes. Cell lysates were prepared, and immunoprecipitation with 880 RNA, Vol.12, No.5 JOBNAME:RNA12#52006PAGE:10OUTPUT:MondayApril315:40:282006 csh/RNA/111792/RNA23396 Downloaded from rnajournal.cshlp.org on February 11, 2023 - Published by Cold Spring Harbor Laboratory Press L22 bindsmultipledomains ofEBER1 anti-L22 antibody (Toczyski and Steitz 1993) was carried out as Haluska,F.G.,Russo,G.,Kant,J.,Andreef,M.,andCroce,C.M.1989. described (Cook et al. 2004). Precipitated RNAs were released Molecular resemblance of an AIDS-associated lymphoma and from the antibody by proteinase K digestion and resolved on an endemicBurkittlymphomas:Implicationsfortheirpathogenesis. Proc.Natl.Acad.Sci.86:8907–8911. 8%ureapolyacrylamidegel.EBER1andEBER2sequenceswere Henle,G.andHenle,W.1966.Immunofluorescence incellsderived detected by Northern blotting using [a-32P]-labeled DNA oligo- fromBurkitt’slymphoma.J.Bacteriol.91:1248–1256. nucleotide probes, EBER 1R152 (59-CCAGCTGGTACTTGACC Henle,G.,Henle,W.,andDiehl,V.1968.RelationofBurkitt’stumor- GAAGAC)andEBER2R134(59-ATTAGAGAATCCTGACTTGCA associated herpes-type virus to infectious mononucleosis. Proc. AATGCTCT). Natl.Acad.Sci.59:94–101. Howe, J.G. and Shu, M.D. 1988. Isolation and characterization of the genes for two small RNAs of herpesvirus papio and their ACKNOWLEDGMENTS comparisonwithEpstein-Barrvirus-encodedEBERRNAs.J.Virol. 62:2790–2798. We thank Kazio Tycowski and Nikolay Kolev for comments on Howe, J.G. and Steitz, J.A. 1986. Localization of Epstein-Barr virus- the manuscript and Enrique De La Cruz, Robert P. Rambo, and encodedsmallRNAsbyinsituhybridization.Proc.Natl.Acad.Sci. TimothyC.Taylorfortheircontributionstoearlierphasesofthis 83:9006–9010. Johansson,B.,Klein,G.,Henle,W.,andHenle,G.1970.Epstein-Barr study. R.M.M-F. is a fellow of the Jane Coffin Childs Fund for virus(EBV)-associatedantibodypatternsinmalignantlymphoma Medical Research. This work was supported by grant CA16038 andleukemia.I.Hodgkin’sdisease.Int.J.Cancer6:450–462. from the NIH. J.A.S. is an investigator of the Howard Hughes Johansson, B., Killander, D., Holm, G., Mellstedt, H., Henle, G., Medical Institute. Henle, W., Klein, G., and Soderberg, G. 1975. Epstein-Barr virus (EBV)-associatedantibodypatternsinrelationtothedeficiencyof Received December 20, 2005; acceptedJanuary 25, 2006. cell-mediatedimmunityinpatientswithHodgkin’sdisease.IARC Sci.Publ.11:237–247. Komano,J.,Maruo,S.,Kurozumi,K.,Oda,T.,andTakada,K.1999. Oncogenic role of Epstein-Barr virus-encoded RNAs in Burkitt’s REFERENCES lymphomacelllineAkata.J.Virol.73:9827–9831. Arrand, J.R. and Rymo, L. 1982. Characterization of the major Koyama, Y., Katagiri, S., Hanai, S., Uchida,K., and Miwa, M. 1999. Epstein-Barr virus-specific RNA in Burkitt lymphoma-derived Poly(ADP-ribose) polymerase interacts with novel Drosophila cells.J.Virol.41:376–389. ribosomal proteins, L22 and l23a, with unique histone-like Arrand, J.R., Young, L.S., and Tugwood, J.D. 1989. Two families of amino-terminalextensions.Gene226:339–345. sequencesinthesmallRNA-encodingregionofEpstein-Barrvirus Le, S., Sternglanz, R., and Greider, C.W. 2000. Identification of two (EBV)correlatewithEBVtypesAandB.J.Virol.63:983–986. RNA-binding proteins associated with human telomerase RNA. Barletta,J.M.,Kingma,D.W.,Ling,Y.,Charache,P.,Mann,R.B.,and Mol.Biol.Cell11:999–1010. Ambinder,R.F.1993.Rapidinsituhybridizationforthediagnosis Lee,S.I.,Murthy,S.C.,Trimble,J.J.,Desrosiers,R.C.,andSteitz,J.A. oflatentEpstein-Barrvirusinfection.Mol.Cell.Probes7:105–109. 1988. FournovelU RNAsareencoded byaherpesvirus. Cell54: Clarke, P.A., Sharp, N.A., and Clemens, M.J. 1990. Translational 599–607. controlbytheEpstein-BarrvirussmallRNAEBER-1.Reversalof Leopardi, R. and Roizman, B. 1996. Functional interaction and thedouble-strandedRNA-inducedinhibitionofproteinsynthesis colocalization of the herpes simplex virus 1 major regulatory inreticulocytelysates.Eur.J.Biochem.193:635–641. proteinICP4withEAP,anucleolar-ribosomalprotein.Proc.Natl. Clarke,P.A.,Schwemmle,M.,Schickinger,J.,Hilse,K.,andClemens,M.J. Acad.Sci.93:4572–4576. 1991.BindingofEpstein-BarrvirussmallRNAEBER-1tothedouble- Leopardi, R., Ward, P.L., Ogle, W.O., and Roizman, B. 1997. stranded RNA-activated protein kinase DAI. Nucleic Acids Res. 19: AssociationofherpessimplexvirusregulatoryproteinICP22with 243–248. transcriptionalcomplexescontainingEAP,ICP4,RNApolymerase Clemens,M.J.andElia,A.1997.Thedouble-strandedRNA-dependent II, and viral DNA requires posttranslational modification by the protein kinase PKR: Structure and function. J. Interferon Cytokine U(L)13proteinkinase.J.Virol.71:1133–1139. Res.17:503–524. Lerner, M.R., Andrews, N.C., Miller, G., and Steitz, J.A. 1981. Two Clemens, M.J., Laing, K.G., Jeffrey, I.W., Schofield, A., Sharp, T.V., small RNAs encoded by Epstein-Barr virus and complexed with Elia,A.,Matys,V.,James,M.C.,andTilleray,V.J.1994.Regulation proteinareprecipitatedbyantibodiesfrompatientswithsystemic oftheinterferon-inducibleeIF-2aproteinkinasebysmallRNAs. lupuserythematosus.Proc.Natl.Acad.Sci.78:805–809. Biochimie76:770–778. Myer, V.E., Lee, S.I., and Steitz, J.A. 1992. Viral small nuclear Cook, H.L., Mischo, H.E., and Steitz, J.A. 2004. The Herpesvirus ribonucleoproteins bind a protein implicated in messenger RNA saimiri small nuclear RNAs recruit AU-rich element-binding destabilization.Proc.Natl.Acad.Sci.89:1296–1300. proteinsbutdonotalterhostAU-richelement-containingmRNA Nakao, A., Yoshihama, M., and Kenmochi, N. 2004. RPG: The levelsinvirallytransformedTcells.Mol.Cell.Biol.24:4522–4533. ribosomal protein gene database. Nucleic Acids Res. 32: D168– Dobbelstein,M.andShenk,T.1995.InvitroselectionofRNAligands D170. fortheribosomalL22proteinassociatedwithEpstein-Barrvirus- Ruf, I.K., Rhyne, P.W., Yang, C., Cleveland, J.L., and Sample, J.T. expressed RNA by using randomized and cDNA-derived RNA 2000.Epstein-BarrvirussmallRNAspotentiatetumorigenicityof libraries.J.Virol.69:8027–8034. Burkitt lymphoma cells independently of an effect on apoptosis. Elia, A., Laing, K.G., Schofield, A., Tilleray, V.J., and Clemens, M.J. J.Virol.74:10223–10228. 1996.Regulationofthedouble-strandedRNA-dependentprotein Ruf, I.K., Lackey, K.A., Warudkar, S., and Sample, J.T. 2005. Pro- kinase PKR by RNAs encoded by a repeated sequence in the tection from interferon-induced apoptosis by Epstein-Barr virus Epstein-Barrvirusgenome.NucleicAcidsRes.24:4471–4478. small RNAs is not mediated by inhibition of PKR. J. Virol. 79: Elia, A., Vyas, J., Laing, K.G., and Clemens, M.J. 2004. Ribosomal 14562–14569. proteinL22inhibitsregulationofcellularactivitiesbytheEpstein- Sharp, T.V., Schwemmle, M., Jeffrey, I., Laing, K., Mellor, H., BarrvirussmallRNAEBER-1.Eur.J.Biochem.271:1895–1905. Proud, C.G., Hilse, K., and Clemens, M.J. 1993. Comparative Glickman,J.N.,Howe,J.G.,andSteitz,J.A.1988.Structuralanalyses analysis of the regulation of the interferon-inducible protein of EBER 1 and EBER2 ribonucleoprotein particles present in kinase PKR by Epstein-Barr virus RNAs EBER-1 and EBER-2 Epstein-Barrvirus-infectedcells.J.Virol.62:902–911. andadenovirusVAIRNA.NucleicAcidsRes.21:4483–4490. www.rnajournal.org 881

Description:
EBER 1, a small noncoding viral RNA abundantly expressed in all cells transformed by Epstein-Barr virus (EBV), has been shown to associate with the
See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.