viral dna tethering domains complement replication...

6
Viral DNA tethering domains complement replication-defective mutations in the p12 protein of MuLV Gag William M. Schneider a,1,2 , Jonathon D. Brzezinski a,b,1 , Sriram Aiyer b , Nirav Malani c , Mercedes Gyuricza a , Frederic D. Bushman c , and Monica J. Roth a,b,3 Departments of a Biochemistry and b Pharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ 08854; and c Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 Edited* by Stephen P. Goff, Columbia University College of Physicians and Surgeons, New York, NY, and approved April 5, 2013 (received for review December 13, 2012) The p12 protein of murine leukemia virus (MuLV) group-specic antigen (Gag) is associated with the preintegration complex, and mutants of p12 (PM14) show defects in nuclear entry or retention. Here we show that p12 proteins engineered to encode peptide sequences derived from known viral tethering proteins can direct chromatin binding during the early phase of viral replication and rescue a lethal p12-PM14 mutant. Peptides studied included seg- ments of Kaposi sarcoma herpesvirus latency-associated nuclear an- tigen (LANA) 123 , human papillomavirus 8 E2, and prototype foamy virus chromatin-binding sequences. Amino acid substitutions in Kaposi sarcoma herpesvirus LANA and prototype foamy virus chro- matin-binding sequences that blocked nucleosome association failed to rescue MuLV p12-PM14. Rescue by a larger LANA peptide, LANA 132 , required second-site mutations that are predicted to re- duce peptide binding afnity to chromosomes, suggesting that ex- cessively high binding afnity interfered with Gag/p12 function. This is supported by confocal microscopy of chimeric p12-GFP fusion constructs showing the reverted proteins had weaker association to condensed mitotic chromosomes. Analysis of the integration-site selection of these chimeric viruses showed no signicant change in integration prole compared with wild-type MuLV, suggesting release of the tethered p12 post mitosis, before viral integration. gammaretroviral vectors | retroviral integration | nuclear retention R etroviral pathogenesis and the utility of retroviruses in gene therapy are greatly inuenced by the requirement for cell di- vision and target-site preferences for integration within host chromosomes. Lentiviruses, such as HIV, are capable of infecting nondividing cells and display a bias toward integrating within ac- tive genes (1). In contrast, infection by gammaretroviruses such as murine leukemia virus (MuLV) requires cell division (2), and in- tegration is biased toward transcriptional start sites (3, 4) and CpG islands (3, 5). In MuLV, the factors responsible for this observed bias in integration target-site selection have not been dened. During a retroviral infection, viral RNA is reverse-transcribed to form double-stranded DNA that is maintained in a preintegration complex (PIC) containing both viral and host proteins. The MuLV PIC includes the viral reverse transcriptase (RT), capsid (CA), integrase (IN), and p12 proteins (68), as well as host factors such as barrier-to-autointegration factor (9). Following reverse transcription, the PIC trafcs to the nucleus, where it associates with chromatin, and ultimately the viral DNA is integrated into host chromosomes, resulting in a stable provirus. For MuLV, the breakdown of the nuclear envelope that occurs during mitosis is necessary for the PIC to gain access to host chromatin; however, the mechanism by which the PIC is retained in the nucleus following mitosis is not known. The MuLV PIC component, p12, functions in both early and late stages of the virus life cycle. The involvement of the p12 protein in late stages of the virus life cycle is attributed to the presence of the late domain (L domain; 31 PPPY 34 ), which functions during viral assembly (10, 11). A role for p12 in early stages of the life cycle was rst demonstrated with work that used alanine-scanning mutagen- esis within the N (PM5, 7) and C terminus (PM13-15) of p12 (11). From this work it was proposed that p12 might have a role in nuclear retention (11). Further investigation demonstrated that mutations in the N terminus of p12 affect very early events in the virus life cycle, possibly through stabilization of the viral core, whereas mutations in the C terminus of p12 may affect chromatin binding (12). Using tagged p12, complexes were detected by immunouorescence in the cytoplasm early after viral infection, and these complexes trafc toward the nucleus and later accumulate adjacent to mitotic chromosomes, supporting a role of p12 in chromatin binding (8). Tethering of viral DNA to host chromatin during mitosis is a common strategy by which DNA viruses maintain stable epi- somal DNA in the nucleus. In addition, lentiviruses, spumaviruses, and retrotransposons (13) all possess mechanisms for tethering viral DNA to host chromatin before integration. For HIV and transposable elements, this tethering function is directly linked to integration. Tethering and targeting of the lentiviral PIC for in- tegration occurs through the interaction of the IN with the host protein lens epithelium-derived growth factor/p75 (14, 15). For the spumavirus prototype foamy virus (PFV), a 13-amino acid se- quence within the group-specic antigen (Gag) C terminus has been shown to bind the H2A/H2B core nucleosome (16), the same ligand bound by the Kaposi sarcoma-associated herpes- virus (KSHV) latency-associated nuclear antigen (LANA) protein (17). Despite all that is known about viral DNA tethers, the molecular tether for MuLV, either for integration or for nuclear retention following cell division, remains elusive. Here we have examined whether the p12 protein of MuLV serves to tether viral DNA to host chromatin. The results of these studies indicate that MuLV containing the p12-PM14 mutation, which displays a defect during the early phase of virus infection, can be restored to replication competence by altering the gene that encodes p12 to include fusions with alternative viral tethering domains. Tethering domains that are capable of complementation include sequences derived from the human papillomavirus 8 Author contributions: W.M.S., J.D.B., S.A., and M.J.R. designed research; W.M.S., J.D.B., S.A., and M.G. performed research; F.D.B. contributed new reagents/analytic tools; W.M.S., J.D.B., S.A., N.M., F.D.B., and M.J.R. analyzed data; and W.M.S., J.D.B., S.A., F.D.B., and M.J.R. wrote the paper. The authors declare no conict of interest. *This Direct Submission article had a prearranged editor. Data deposition: The sequences reported in this paper have been deposited in the Na- tional Center for Biotechnology Information Sequence Read Archive (project accession number SRP021184). See Commentary on page 9195. 1 W.M.S. and J.D.B. contributed equally to this work. 2 Present address: Center for the Study of Hepatitis C, Laboratory of Virology and Infec- tious Disease, The Rockefeller University, New York, NY 10021. 3 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1221736110/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1221736110 PNAS | June 4, 2013 | vol. 110 | no. 23 | 94879492 MICROBIOLOGY SEE COMMENTARY

Upload: others

Post on 05-Oct-2020

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Viral DNA tethering domains complement replication ...bushmanlab.org/assets/pdf/publications/schneider_2013.pdf · Viral DNA tethering domains complement replication-defective mutations

Viral DNA tethering domains complementreplication-defective mutations in thep12 protein of MuLV GagWilliam M. Schneidera,1,2, Jonathon D. Brzezinskia,b,1, Sriram Aiyerb, Nirav Malanic, Mercedes Gyuriczaa,Frederic D. Bushmanc, and Monica J. Rotha,b,3

Departments of aBiochemistry and bPharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway,NJ 08854; and cDepartment of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104

Edited* by Stephen P. Goff, Columbia University College of Physicians and Surgeons, New York, NY, and approved April 5, 2013 (received for reviewDecember 13, 2012)

The p12 protein of murine leukemia virus (MuLV) group-specificantigen (Gag) is associated with the preintegration complex, andmutants of p12 (PM14) show defects in nuclear entry or retention.Here we show that p12 proteins engineered to encode peptidesequences derived from known viral tethering proteins can directchromatin binding during the early phase of viral replication andrescue a lethal p12-PM14 mutant. Peptides studied included seg-ments of Kaposi sarcoma herpesvirus latency-associated nuclear an-tigen (LANA)1–23, human papillomavirus 8 E2, and prototype foamyvirus chromatin-binding sequences. Amino acid substitutions inKaposi sarcoma herpesvirus LANA and prototype foamy virus chro-matin-binding sequences that blocked nucleosome associationfailed to rescue MuLV p12-PM14. Rescue by a larger LANA peptide,LANA1–32, required second-site mutations that are predicted to re-duce peptide binding affinity to chromosomes, suggesting that ex-cessively high binding affinity interfered with Gag/p12 function.This is supported by confocal microscopy of chimeric p12-GFP fusionconstructs showing the reverted proteins had weaker association tocondensed mitotic chromosomes. Analysis of the integration-siteselection of these chimeric viruses showed no significant changein integration profile compared with wild-type MuLV, suggestingrelease of the tethered p12 post mitosis, before viral integration.

gammaretroviral vectors | retroviral integration | nuclear retention

Retroviral pathogenesis and the utility of retroviruses in genetherapy are greatly influenced by the requirement for cell di-

vision and target-site preferences for integration within hostchromosomes. Lentiviruses, such as HIV, are capable of infectingnondividing cells and display a bias toward integrating within ac-tive genes (1). In contrast, infection by gammaretroviruses such asmurine leukemia virus (MuLV) requires cell division (2), and in-tegration is biased toward transcriptional start sites (3, 4) and CpGislands (3, 5). In MuLV, the factors responsible for this observedbias in integration target-site selection have not been defined.During a retroviral infection, viral RNA is reverse-transcribed to

form double-stranded DNA that is maintained in a preintegrationcomplex (PIC) containing both viral and host proteins. The MuLVPIC includes the viral reverse transcriptase (RT), capsid (CA),integrase (IN), and p12 proteins (6–8), as well as host factors such asbarrier-to-autointegration factor (9). Following reverse transcription,the PIC traffics to the nucleus, where it associates with chromatin,and ultimately the viral DNA is integrated into host chromosomes,resulting in a stable provirus. For MuLV, the breakdown of thenuclear envelope that occurs during mitosis is necessary for the PICto gain access to host chromatin; however, the mechanism by whichthe PIC is retained in the nucleus following mitosis is not known.TheMuLV PIC component, p12, functions in both early and late

stages of the virus life cycle. The involvement of the p12 protein inlate stages of the virus life cycle is attributed to the presence of thelate domain (L domain; 31PPPY34), which functions during viralassembly (10, 11). A role for p12 in early stages of the life cycle was

first demonstrated with work that used alanine-scanning mutagen-esis within the N (PM5, 7) and C terminus (PM13-15) of p12 (11).From this work it was proposed that p12might have a role in nuclearretention (11). Further investigation demonstrated that mutationsin theN terminus of p12 affect very early events in the virus life cycle,possibly through stabilization of the viral core, whereas mutationsin the C terminus of p12 may affect chromatin binding (12). Usingtagged p12, complexes were detected by immunofluorescencein the cytoplasm early after viral infection, and these complexestraffic toward the nucleus and later accumulate adjacent to mitoticchromosomes, supporting a role of p12 in chromatin binding (8).Tethering of viral DNA to host chromatin during mitosis is

a common strategy by which DNA viruses maintain stable epi-somal DNA in the nucleus. In addition, lentiviruses, spumaviruses,and retrotransposons (13) all possess mechanisms for tetheringviral DNA to host chromatin before integration. For HIV andtransposable elements, this tethering function is directly linked tointegration. Tethering and targeting of the lentiviral PIC for in-tegration occurs through the interaction of the IN with the hostprotein lens epithelium-derived growth factor/p75 (14, 15). For thespumavirus prototype foamy virus (PFV), a 13-amino acid se-quence within the group-specific antigen (Gag) C terminus hasbeen shown to bind the H2A/H2B core nucleosome (16), thesame ligand bound by the Kaposi sarcoma-associated herpes-virus (KSHV) latency-associated nuclear antigen (LANA) protein(17). Despite all that is known about viral DNA tethers, themolecular tether for MuLV, either for integration or for nuclearretention following cell division, remains elusive.Here we have examined whether the p12 protein of MuLV

serves to tether viral DNA to host chromatin. The results of thesestudies indicate that MuLV containing the p12-PM14 mutation,which displays a defect during the early phase of virus infection,can be restored to replication competence by altering the gene thatencodes p12 to include fusions with alternative viral tetheringdomains. Tethering domains that are capable of complementationinclude sequences derived from the human papillomavirus 8

Author contributions: W.M.S., J.D.B., S.A., and M.J.R. designed research; W.M.S., J.D.B., S.A., andM.G. performed research; F.D.B. contributed new reagents/analytic tools; W.M.S., J.D.B., S.A.,N.M., F.D.B., andM.J.R. analyzeddata; andW.M.S., J.D.B., S.A., F.D.B., andM.J.R.wrote thepaper.

The authors declare no conflict of interest.

*This Direct Submission article had a prearranged editor.

Data deposition: The sequences reported in this paper have been deposited in the Na-tional Center for Biotechnology Information Sequence Read Archive (project accessionnumber SRP021184).

See Commentary on page 9195.1W.M.S. and J.D.B. contributed equally to this work.2Present address: Center for the Study of Hepatitis C, Laboratory of Virology and Infec-tious Disease, The Rockefeller University, New York, NY 10021.

3To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1221736110/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1221736110 PNAS | June 4, 2013 | vol. 110 | no. 23 | 9487–9492

MICRO

BIOLO

GY

SEECO

MMEN

TARY

Page 2: Viral DNA tethering domains complement replication ...bushmanlab.org/assets/pdf/publications/schneider_2013.pdf · Viral DNA tethering domains complement replication-defective mutations

(HPV8) E2 protein (18, 19) as well as the chromatin bindingsequences (CBSs) from both the KSHV LANA and PFV Gagproteins. We found that the integration profiles of the infectiousp12 chimeric viruses derived from KSHV, PFV, and HPV8 werenot significantly altered from the integration profile for MuLV,which implies the DNA tethering function provided by p12 is fornuclear localization/retention during mitosis and does not in-fluence genomic targeting of the integration event.

ResultsDesign of MuLV Chimeric p12 Variants. Fig. 1A outlines the aminoacid sequence of MuLV Gag p12, highlighting regions of impor-tance. We hypothesized that in addition to the L domain, whichvaries in position between murine and avian viruses and lentiviruses(10, 11), the p12 protein may also harbor a domain that contributesa tethering function. We chose to study the MuLV p12-PM14mutation in complementation assays due to the homology of theRXR residues to the CBSs of LANA and PFV. The p12-PM14isolate initially reported (11) encodes Ile at position 63, and is theprototype parental p12 sequence used in these studies, exceptwhere noted. Previous studies have identified regions of p12 at theN terminus (5PS) (20) and the central region (substituting 49G) thatare tolerant to insertions (21). These positions were used as landingsites for the insertion of alternative tethering domains.A panel of transport and tethering domains was analyzed for

functional complementation of MuLV p12-PM14 (Table 1). Thep12 mutations were analyzed using two assays. In the first, the p12constructs were engineered into a full-length provirus, and viralreplication was measured through multiple rounds of infection inD17 cells rendered permissive for MuLV infection (22). Cellswere transfected with proviral DNA and virus production wasmeasured by the release of RT activity into the media over time(Figs. 1B, 2B, 3A, and 4B) (23). In the second assay, individualp12 constructs were fused to GFP, and their subcellular locali-zation was determined by confocal microscopy in the absence ofother viral proteins (Figs. 1 C and D, 2C, 3B, and 4C).

Viral Tethering Factors Complement the p12-PM14Mutation in a Position-Dependent Fashion and Restore Chromosomal Tethering. Results of theviral spread assay measuring the function of the p12 fusions are

shown in Fig. 1B. Supernatants collected from cells that weretransfected with the parental virus were RT-positive within 10 d,whereas cells that were mock-transfected remained RT-negativefor the duration of the experiment. Cells transfected with MuLVp12-PM14 DNA or variants encoding substitutions of the argi-nine residues within PM14 (R66A/R68A) remained RT-negativefor the duration of the experiment (40 d).Insertion of the LANA1–23 sequence into MuLV p12-PM14 at

the N terminus of p12 (p12-N-LANA1–23-PM14) did not result indetectable virus production, but inserting this sequence withinthe central region of p12 generated viable virus (p12-LANA1–23-PM14). This chimeric virus showed delayed growth kinetics rel-ative to MuLV, with the detection of RT activity first appearingon day 20. Proviral DNA from infected cells was sequenced,confirming that the LANA1–23 insertion was maintained and noadditional second-site mutations accumulated within p12.Similar to KSHV LANA1–23 (17), the PFV CBS is reported to

tether to the core nucleosome (16). Insertion of a 13-aa region ofthe PFV Gly-Arg II domain into the central region of p12 wasalso capable of rescuing MuLV p12-PM14, with even faster ki-netics than the LANA1–23 insertion (Fig. 1B).Like KSHV and PFV, a chromatin tethering function is also

present in HPV8. For HPV8 the chromatin tethering function iscontained within the C-terminal domain (CTD) of the E2 pro-tein, which works in concert with the central hinge region.Rather than inserting the large CTD of E2 along with the hingeregion, the hinge region itself was tested for the ability to com-plement MuLV p12-PM14. The E2 hinge region regulatesbinding of the E2 CTD through phosphorylation, prompting us to

Fig. 1. Summary of MuLV passage of virus bearing p12 mutations and functional complementation. (A) Schematic of MuLV p12. Sequence of the MuLV p12protein with key features highlighted. Triangle, position of insertions, N-terminal (PS) (yellow box) and central insertion site (insertions replace boxed G) (redbox); blue box, late assembly sequence; turquoise box, Δ50–64; asterisk, phosphorylated S61; and green box, PM14 region (mutated to Ala). (B) Viral passage inD17pJET cells measured by RT activity and scored for the day the culture was RT-positive. SEs are indicated (n ≥ 3). Δ, p12 deletion Δ50–64. The asterisk indicatesvirus replicating contains second-site revertants. (C) Tethering of p12-PM14-GFP constructs to mitotic chromatin in D17 cells. Ratios of GFP overlapping theDNA of mitotic D17 cells (tethered GFP) intensity:cytoplasmic GFP intensity were averaged for each construct (n > 10) with SEs shown. Student t tests wereperformed against GFP control; *P < 0.001. (D) Images representing each construct. Images with a tethered ratio below 1.0 show GFP excluded from themitotic chromatin, whereas images with ratios above 1.0 show significant overlap of GFP with mitotic chromatin. (Scale bars, 10 μm.)

Table 1. p12 inserts

Insert Sequence

KSHV LANA1–23 MAPPGMRLRSGRSTGAPLTRGSC

KSHV LANA1–32 MAPPGMRLRSGRSTGAPLTRGSCRKRNRSPER

KSHV NLS24–32 RKRNRSPER

SV40 TAg NLS PPKKKRKV

PFV Gag534–546 QGGYNLRPRTYQP

HPV8 E2240–255 QTETKGRRYGRRPSSR

9488 | www.pnas.org/cgi/doi/10.1073/pnas.1221736110 Schneider et al.

Page 3: Viral DNA tethering domains complement replication ...bushmanlab.org/assets/pdf/publications/schneider_2013.pdf · Viral DNA tethering domains complement replication-defective mutations

analyze chimeric MuLV p12-PM14 constructs in the presence orabsence of the phosphorylation domain of p12 (Δ50–64 region; Fig.1) (24). The results demonstrated that the hinge region of E2 isunable to rescue MuLV p12-PM14. However, when the p1250–64region was deleted (p12-E2-Δ-PM14), viable virus was detectedafter ∼26 d (discussed below in detail; Fig. 4 and Fig. S1). Deletionof p1250–64 had no effect on the ability of the tethering domainsfrom either the PFV Gag (p12-PFV-Δ-PM14) or LANA1–23 (p12-LANA1–23-Δ-PM14) insertions to rescue MuLV p12-PM14 (Fig.S2). Viral protein analysis of all viable viruses indicated that pro-teolytic processing of Gag was similar to WT virus (Fig. S3).We next examined the distribution of p12-GFP chimeras on

mitotic chromosomes in the absence of other viral proteins. D17cell lines stably expressing the p12-GFP constructs were gener-ated and analyzed by confocal microscopy (Fig. 1 C and D). Allp12-GFP chimeras were analyzed by Western blot to confirm thatthe full-length protein was stable and expressed (Fig. S4). Imageswere quantified as the ratio of tethered GFP intensity overlappingthe Hoechst-stained condensed mitotic DNA compared with cy-toplasmic GFP intensity. Student t test was used to compare theratio of the chimeric constructs with that of GFP. In this system,the GFP alone is not tethered to mitotic chromosomes, whereasthe parental p12 shows significant tethering (ratio = 1.16, P <0.001), which is lost in the presence of either the PM14 or R66A/R68A mutations (Fig. 1C). Introduction of the LANA1–23 CBSshowed a marked tethering to the mitotic chromosomes (ratio =4.06, P < 0.001) independent of the p1250–64 region (Δ) (Fig. 1 C

and D and Fig. S2 B and C). Similarly, insertion of the 534PFV546CBS into p12-PM14 showed measurable tethering to mitoticchromosomes (ratio = 1.22, P < 0.001) independent of the p1250–64region (Fig. 1 C and D and Fig. S2).These results indicate that known tethering domains from var-

ious viruses are capable of complementing a defect in an earlyphase of MuLV infection when fused to p12 and that the positionof insertion is critical for function. Factors that could alter thetransport of the PIC to the nucleus did not overcome the p12-PM14 defect. Insertion of nuclear localization sequences (NLS),including the classical SV40 T antigen (TAg) NLS and the non-classical KSHV LANA NLS (LANA24–32), did not rescue MuLVp12-PM14, and neither showed tethering to the mitotic chromatin(Fig. S2). To gain better insight as to the mechanism by whichthe alternative tethering domains rescued MuLV p12-PM14, wetested the effect of substitutions of key amino acids within thesetethering domains.

Complementation by PFV and LANA1–23 CBSs Requires Amino AcidsAssociated with Nucleosome Tethering. The 534PFV546 CBS withinMuLV p12-PM14 virus was mutated and tested to determinewhether it impaired viral rescue (Fig. 2A). Our results indicate thattwo arginine residues (R7 and R9) within this insert are essentialfor the functional complementation of MuLV p12-PM14; R9could not be substituted with Lys (R9K), indicating that a positivecharge alone is not sufficient for function (Fig. 2B). These resultsagree with the mutations resulting in the loss of PFV tetheringpreviously reported for 539LRP→AAA and 542RTY→AAA (16).Introduction of the 534PFV546 into p12 showed marked teth-

ering to the mitotic chromosomes. Tethering, however, was lostin constructs containing mutations in R7 and R9 (PFV R7A,PFV R9K, and PFV 9RTY11→AAA), providing a direct corre-lation between tethering activity and complementation of p12-PM14 (Fig. 2 B and C).The KSHV LANA1–23 insert was able to rescue p12-PM14

(Fig. 1B). Arg9 of LANA1–23 forms multiple hydrogen bonds toH2A, and mutation of 8LRS10 to Ala resulted in the loss ofchromosomal tethering (17). Substitution of R9A/R12A withinthe LANA1–23 peptide in the context of MuLV p12-PM14 didnot yield viable virus (Fig. 2B), and showed a corresponding lossof tethering to mitotic chromosomes (Fig. 2C).

Reversion Events Optimize the Level of Tethering Required for ViralReplication. Results with chimeric viruses containing the largerLANA1–32 encoding the putative NLS were distinct from thoseobserved for LANA1–23. Only one of four independent assays ofp12-LANA1–32-PM14 chimeras yielded infectious virus (day 41).

Fig. 2. Mutational analysis of MuLV p12-PFV-PM14 and p12-LANA1–23-PM14 inserts. (A) Schematic of the p12-PM14 mutation with the 13-aa PFVand 23-aa LANA sequences indicated. Numbering corresponding to the PFVGag is shown in parentheses. Positions in red, when mutated, result in theloss of complementation of the viral construct bearing the p12-PM14 mu-tant. (B) Viral passage in D17pJET cells measured by RT activity. SEs are in-dicated (n ≥ 3). (C) Tethering of p12-PFV-PM14-GFP constructs to mitoticchromatin in D17 cells. Ratios of tethered GFP intensity:cytoplasmic GFP in-tensity were averaged for each construct (n > 10) with SEs shown. Constructswith a ratio above 1.0 correlate with live virus in B. Student t tests wereperformed against GFP control; *P < 0.001.

Fig. 3. Reversion of the MuLV p12–LANA1–32-PM14 insert. (A) Schematic ofp12-LANA1–32-PM14 insert. REV, revertant isolated after viral passage; box,reported NLS arginine-rich region. The day the culture was RT-positive is in-dicated with SE (n = 3). (B) Confocal images of each p12-LANA1–32-PM14 con-struct. Ratios of tethered GFP intensity:cytoplasmic GFP intensity were averagedfor each construct (n > 10) with SEs shown; *P < 0.001. (Scale bars, 10 μm.)

Schneider et al. PNAS | June 4, 2013 | vol. 110 | no. 23 | 9489

MICRO

BIOLO

GY

SEECO

MMEN

TARY

Page 4: Viral DNA tethering domains complement replication ...bushmanlab.org/assets/pdf/publications/schneider_2013.pdf · Viral DNA tethering domains complement replication-defective mutations

Sequence analysis of the integrated p12-LANA1–32-PM14 pro-viral DNA identified a single-point mutation, R9K; the reversionevent is indicative of a low level of viral passage. Reconstitutionwithin a MuLV provirus and subsequent infection studies in-dicated that this revertant had the same replication kinetics as theviral derivative containing the LANA1–23 insert (day 20) (Fig. 3A).Mutation of R9A resulted in replication kinetics faster than thep12-PM14 bearing either the LANA1–23 or LANA1–32-R9K inserts(Fig. 3A). Selection for R9K was of interest because Arg9 wasidentified as critical for nucleosome binding for both LANA1–23and LANA1–32 and inactivated complementation of the LANA1–23insert (Fig. 2B). We therefore predicted that the virus had selectedfor chimeric p12 derivatives with decreased nucleosome affinity,and this was directly analyzed in the context of p12-GFP fusions.Fig. 3B shows the results of the mitotic tethering of the LANA

inserts within the p12-GFP constructs. The p12-LANA1–32-PM14-GFP construct showed strong tethering to mitotic chromosomes,with a tethered GFP intensity:cytoplasmic GFP intensity ratio of7.76 (Fig. 3B). Despite the strong tethering, virus bearing thisinsert was not viable and selected the second-site mutation R9Kto restore viability as described above. Analysis of this R9Krevertant by confocal microscopy indicated a marked decrease inchromosomal tethering, with a decrease in ratio from 7.76 to 1.22.Additionally, the p12-LANA1–32-R9A-PM14 virus, which was the

fittest of the LANA1–32 viral constructs, had the lowest tetheringratio at 1.14 (Fig. 3B). These results indicate that tight tetheringto mitotic chromosomes is detrimental to MuLV infection andthat mutations are selected that weaken the association with themitotic chromosomes to allow for productive infection.To confirm that colocalization of the GFP signal with chromatin

was indeed due to binding, fluorescence recovery after photo-bleaching (FRAP) was performed on mitotic cells (Fig. S5). Thetime of recovery for all p12 chimeras tested was greater than freeGFP (0.2 s). Specifically, the time to 50% recovery (t1/2) for p12-GFP and the p12-PM14-GFP constructs bearing LANA1–23 andLANA1–32 was 0.5, 1.4, and 5.4 s, respectively. These results sup-port the model that p12-GFP chimeras are bound to chromatin.The increased FRAP time signifies the time required for chimericGFP dissociation, reflecting their increased binding affinity.The direct association of p12-LANA1–32-PM14-GFPwith histone

H2B was confirmed by coimmunoprecipitation. Incorporation ofthe R9K mutation into the extended LANA1–32 region decreasedH2B association below detectable levels (Fig. S6).

Complementation with the HPV8 E2 Hinge Region240–255 Identifiesa Regulatory Sequence Within p12. Insertion of the HPV8 E2hinge region240–255 (numbered 1–16; Fig. 4) into MuLV p12-PM14 did not complement the PM14 mutation (Fig. 4B).Surprisingly, deletion of the central region of p12 from aminoacids 50–64 in p12-E2-PM14 resulted in a viral derivative thatproduced a number of independent second-site mutationsduring viral passage. Ten second-site mutations have been identi-fied; reconstruction of eight individual mutations into the parentalproviral construct (p12-E2-Δ50–64-PM14) confirmed that the repli-cation of the proviral constructs was restored to near WT levels(Fig. 4B and Fig. S1D) and that mutations were stable. The E2second-site mutations can be grouped into three regions: Two aredefined by the deletion mutations ΔETKGRRY and ΔRRPSS,and the third lies outside the E2 insertion, at p12 R71Q.Parallel studies examined the effect of passaging virus bearing

Δ50–64. Interestingly, a second-site mutation, G69R, was isolatedfrom p12-Δ50–64, which created a basic patch of RRRR. Thisresulted in a virus that replicated similarly to that encoding theparental p12 (Fig. 4B). These results indicate that there aresimilarities in the second-site mutations obtained from MuLVbearing the p1250–64 deletion, regardless of whether the E2-PM14insert is present.Analysis of the p12-E2-GFP constructs supports the hypothesis

that MuLV requires weak tethering to mitotic chromosomes for vi-ability (Fig. 4C and Fig. S1 E and F). Comparison of the p12-E2-PM14-GFP (Fig. 4C) construct with a construct containing the p12-Δ50–64 deletion (p12-E2-Δ50–64-PM14-GFP; Fig. 4C) reveals thatremoving amino acids 50–64 activates tethering to the mitotic chro-mosomes. Between 10 and 30 images were quantified for the GFPtethering ratio of p12-E2-PM14-GFP (0.98) and p12-E2-Δ50–64-PM14-GFP (1.19), and the measured difference between the con-structs is highly significant (P< 0.001). Interestingly, all eight second-site mutations selected by the virus decrease the level of tetheringfrom theparental p12-E2-Δ50–64-PM14 (Fig. 4C andFig. S1E andF).

Integration Sites Used by Virus Bearing p12 Insertions Do Not Differfrom MuLV with Respect to Transcriptional Start Sites and CpGIslands. To characterize the integration-site profile of MuLV bear-ing p12 chimeras in human cells, replication competent chimericvirus was transfected into 293 cells expressing the ecotropic MuLVreceptor, murine cationic amino acid transporter-1 (293mCAT).Following viral passage, genomic DNA was isolated and between1,100 and 2,200 unique integration sites were mapped for each chi-meric virus (Table S1). MuLV integration sites are enriched neartranscription start sites and CpG islands, whereas FV and HIV in-tegration sites are not. The integration-site distribution of theMuLVp12-PM14 virus encoding PFV (p12-PFV-PM14), LANA (p12-LANA1–23-PM14 and p12-LANA1–32-R9K-PM14), and E2 inserts

Fig. 4. Functional analysis of MuLV p12-E2-PM14 and p12-Δ50–64 inserts. (A)(Top) Schematic of p12-Δ50–64 mutation. Sequence of the PM14 region isshown as well as the p12-Δ50–64-G69R revertant. (Middle and Bottom) Sche-matic of p12-E2-PM14 mutations. The 16-aa HPV8 E2 hinge sequence is in-dicated with HPV8 E2 numbering in parentheses. Positions where individualsecond-site mutations have been identified are highlighted in red and thedeletions are underlined. The positions of the 70RR71 C-terminal to PM14 andthe revertant 70RQ71 are indicated. (B) Passage of virus transiently expressedin D17pJET cells. The day the culture was RT-positive is indicated with SE (n ≥3). Δ, p12 deletion Δ50–64; asterisk, parental construct from which revertantswere isolated. (C) Tethering of p12-E2-Δ50–64-PM14-GFP and p12-Δ50–64-GFPconstructs to mitotic chromatin in D17 cells. Ratios of tethered GFP intensity:cytoplasmic GFP intensity were averaged for each construct (n > 10) with SEsshown. Constructs with a ratio above 1.0 correlate with live virus in B. Studentt tests were performed against GFP control; *P < 0.001.

9490 | www.pnas.org/cgi/doi/10.1073/pnas.1221736110 Schneider et al.

Page 5: Viral DNA tethering domains complement replication ...bushmanlab.org/assets/pdf/publications/schneider_2013.pdf · Viral DNA tethering domains complement replication-defective mutations

(p12-E2-K5E-Δ-PM14 and p12-E2-ΔRRPSS-Δ-PM14) maintainedtheir bias toward integrationat transcriptional start sites (Fig. 5A) andCpG islands (Fig. 5B). Statistical comparisons over many forms ofgenomic annotation are shown in Fig. S7. Although individual sta-tistical variations exist between sets, the tethering of p12 associatedwith mitotic chromosomes during the infectious entry cycle did notalter the overall integration profile of MuLV.

DiscussionIn this study, we show that the insertion of amino acid sequencesknown to be required for tethering viral DNA to host chromatin canfunctionally rescue a replication-defective MuLV bearing a lethalmutation in p12. A key feature that impacts whether a given teth-ering domain is capable of rescue is the binding affinity to mitoticchromosomes. Second-site mutations were identified that reducebinding affinity; however, mutations that are predicted to eliminatebinding to chromosomes are incapable of rescue; this indicates thatthe ability to bind with optimized intermediate affinity to mitoticchromosomes is required for p12 function. In contrast to the in-sertion of tethering domains, the insertion of nuclear localizationsequences within p12 was unable to rescue MuLV p12-PM14. Ourresults indicate that the C terminus of the p12 protein of MuLVfunctions to retain the PIC within the nucleus after the reformationof the nuclear envelope that occurs at the end of mitosis. Further-more, the mitotic chromatin tethering mediated by p12 does notinfluence integration target-site selection directly.The infectious retroviral pathway involves orchestration of mul-

tiple viral and host proteins through the reverse-transcription andpreintegrative complexes. Recent findings indicate that the N ter-minus of p12 plays a role in capsid core disassembly (12).Our resultscorrelate the function of the p12 C terminus to mitotic chromatintethering (12, 25). Live-cell imaging indicates p12-labeled PICstraffic and tether to the mitotic chromatin after nuclear envelopebreakdown, and the p12-PM14 mutation ablates tethering (25).Mutant p12 can influence CA release from PICs in mitotic cells(25). The results of this study indicate that the optimized weaktethering of p12 chimeras selected by viral passage does not in-fluence the subsequent integration events coordinated by the INprotein. These results support previous data demonstrating thatIN is the dominant factor in defining target-site selection (26).The p12 protein has functions in both early and late stages of the

life cycle, and must therefore balance both incoming and outgoingviral pathways. Reversion events selecting for improved virus via-bility can optimize either stage of the viral life cycle. Parallel studiesgenerating a related p12-LANA1–31-PM14 indicated low viral titerand reduced assembly of viral particles within a p12-LANA1–31background (25). Thus, for the p12-LANA1–32-PM14 construct,second-sitemutations can be selected that improve the localization

or assembly of Gag precursor Pr65 at the plasma membrane andconcomitantly decrease binding to the mitotic chromosomes.The LANA NLS is proposed to function as part of a larger

lysine-arginine–rich bipartite NLS (27). p12-LANA1–32-PM14containing the NLS resulted in an increased level of chromosomaltethering. This suggests a role for arginine-rich sequences in bothnuclear retention and chromosome binding. In addition, arginine-rich motifs have been shown to function as RNA binding motifs(28). It is therefore worth noting that both the PM14 and theadjacent PM15 mutations lie within an arginine-rich region that ishighly conserved among retroviral p12 proteins (24). Glycine-arginine–rich regions are also present in viral proteins with anestablished role in chromosome tethering, including the DNAvirus proteins Epstein–Barr virus EBNA1, KSHV LANA, HPVE2, and the retroviral PFV Gag protein (16, 18, 19, 29). The roleof these glycine-arginine–rich regions is unclear.In addition to a glycine-arginine–rich region, the DNA virus

proteins contain a conserved RXXS phosphorylation motif. AnRXXS motif is present within the HPV8 E2 hinge region (19);however, this motif was not essential for complementation ofp12-E2-PM14. In fact, the motif was deleted through viral pas-sage, resulting in a mutant virus, ΔRRPSS, with increased fitness.Although the p12 protein of MuLV lacks an RXXS motif, al-ternative phosphorylation sites within p12 have been identified:S61 (within the Δ50–64 region) and S65 (within PM14). Repli-cation of p12-Δ50–64-G69R, lacking the S61 phosphorylation site,indicates that p12 phosphorylation per se may not be essential.The fact that other tethering domains are capable of restoring

a mutant p12 function provides evidence that the wild-type p12functions in tethering to chromosomes. The collection of com-pensatory mutations in constructs bearing E2 as well as in p12-Δ50–64-G69R reveals a requirement for Arg-rich sequences, in-dicating that the putative host factor could be either protein orRNA (30). The HPV8 E2 hinge region provides a fruitful res-ervoir to facilitate second-site reversions capable of compensat-ing for the PM14 mutation, indicating that amino acids derivedfrom E2 do not necessarily rescue the p12-PM14 mutation in asequence-specific fashion. Minimally, the results of these studiesindicate that tethering to the core nucleosome H2A/H2B throughp12-PM14 fused to the tethering domains of PFV and KSHVLANA proteins is sufficient to restore viral infectivity.The results of this study highlight a role for the MuLV p12

protein in nuclear retention following mitosis using a comparativevirology approach. These results further demonstrate the plasticityof the retroviral genomes. This has been demonstrated previouslywith the late domain, and with this work we report that a similarsituation can be found with tethering functions. For MuLV, the p12protein appears to be the landing pad of multiple accessory func-tions. Further defining the partners involved for tethering MuLV

Fig. 5. Proportion of integration sites surrounding transcription start sites and CpG islands for MuLV engineered to contain the LANA and PFV CBSs, and an E2regulatory region. (A) Percentages of integration sites are plotted with respect to the distance from the annotated transcription start sites. (B) Percentages ofintegration sites are plotted with respect to the distance from the midpoint of the nearest CpG islands. (Inset) Datasets used in the analysis; FV fibroblast (37),HIV-1 (38), MuLV (ref. 39 and this work), MuLV p12-M63, MuLV p12-PFV-PM14, p12-LANA1–23-PM14, p12-LANA1–32-R9K-PM14, p12-E2-K5E-Δ-PM14, and p12-E2-ΔRRPSS-Δ-PM14 (summarized in Table S1). The profiles of the MuLV p12-PM14 chimeras maintained the bias to transcriptional start sites and CpG islands.

Schneider et al. PNAS | June 4, 2013 | vol. 110 | no. 23 | 9491

MICRO

BIOLO

GY

SEECO

MMEN

TARY

Page 6: Viral DNA tethering domains complement replication ...bushmanlab.org/assets/pdf/publications/schneider_2013.pdf · Viral DNA tethering domains complement replication-defective mutations

p12 to mitotic chromosomes and investigating the mechanism/regulation of release remain key areas of interest to this field.

Materials and MethodsPlasmids. pNCA-C encoding the complete Moloney-MuLV provirus was used asthe template to clone the p12 mutants (31). Inserts replaced glycine-49 of p12and were cloned into pNCA-C with overlapping PCR and the EcoRI and XhoIrestriction sites. Mutant p12 constructs were then cloned into the GFP mam-malian expression vector GFP-IRES-Puro (pGIP) (32) with primers flanking p12that introduced an N-terminal BamHI restriction site, ATG start codon, and C-terminal (GGGGS)3 linker to link to GFP. GFP was amplified with primers thatadded the (GGGGS)3 linker to the N terminus and deleted the ATG start codonand included the C-terminal NotI restriction site. Overlapping PCRwas used tocombine the p12 andGFP fragments, and the insert was cloned back into pGIPwith the BamHI and NotI restriction sites. PCR reactions were performedwithKOD polymerase (Novagen) and digests with New England BioLabsenzymes. Oligonucleotide primer sequences are listed in Dataset S1.

Cell Culture. All cells were cultured at 37 °C, 5% CO2 in 4.5 g/L D-glucose,L-glutamine (+) DMEM (Gibco) supplemented with 10% (vol/vol) FBS(Atlanta Biologicals) and 1% (vol/vol) antimycotic/antibiotic (Gibco).

DEAE dextran transfection in canine D17 cells stably expressing the eco-tropic M-MuLV receptor mCAT (D17pJET cells) was as previously describedusing 250 ng proviral DNA (33). Viral replication kinetics were monitoredusing an RT assay performed on the culture supernatants (23). All assayswere performed minimally in three independent experiments.

p12-GFP constructs were expressed in D17 cells. 293T cells stably expressingthe MuLV Gag-Pol proteins (293T CeB cells) (34) were used to generate virusto transduce the D17 cells with the pGIP vectors (32). A CaPO4 transfectionkit (Stratagene; Mammalian Transfection Kit) was used for overnighttransfection of 1.5 μg pHIT-G plasmid expressing the vesicular stomatitisvirus G protein (VSV-G) (35) and 0.5 μg pGIP at 37 °C in 2.5% CO2. Cells were

then treated with 10 mM Na butyrate for 6 h (36), and the medium waschanged and virus was collected 24 h later. D17 cells were infected with virusfor 3 h in the presence of 8 μg/mL polybrene and, after 24 h, selected inmedia containing 2.5 μg/mL puromycin.

Confocal Microscopy. D17 cells expressing the p12-GFP constructs were imagedlive on a Zeiss LSM 510META confocal microscopewith a 63×water immersionobjective at the Robert Wood Johnson Medical School Confocal and Elec-tronic Imaging Center. For some images, subconfluent dishes were treatedwith 0.45 ng/mL nocodazole in medium for 8–10 h and then released fromcell-cycle arrest with fresh medium containing Hoechst 33258 DNA stain(Sigma; 14530) for 30 min. Cells were imaged in MatTek glass-bottom dishes(P35G-1.5-14-C). All cells imaged in metaphase were quantified in Matlab(MathWorks). The area of a single cell was defined for each image excludingsignals outside this domain. Mitotic chromatin was defined as any HoechstDNA staining signal 50% above background. The GFP signal was classified asoverlapping the mitotic chromatin signal (tethered) or as cytoplasmic. Thefinal tethered GFP intensity:cytoplasmic GFP intensity ratio was calculated.

Integration Target-Site Selection Analysis. Genomic DNA containing viralintegrants from infected 293mCAT cells was isolated, fragmented, repaired,and ligated with linkers for 454 pyrosequencing following protocols de-scribed in SI Materials and Methods.

ACKNOWLEDGMENTS. We thank Dirk Moore (University of Medicine andDentistry of New Jersey School of Public Health) for assistance in the statisticalanalysis, Sheyum Syed (The Rockefeller University) for the program to quantifythe confocal microscopy, and Christopher Warren and Roland Felkner for tech-nical assistance. This work was supported by National Institutes of Health (NIH)Grants R01 GM070837 and GM088808 (to M.J.R.) and AI52845 and AI082020(to F.D.B.). J.D.B. was supported by NIH Training Grant T32 GM008360.

1. Schröder ARW, et al. (2002) HIV-1 integration in the human genome favors activegenes and local hotspots. Cell 110(4):521–529.

2. Roe T, Reynolds TC, Yu G, Brown PO (1993) Integration of murine leukemia virus DNAdepends on mitosis. EMBO J 12(5):2099–2108.

3. Wu X, Li Y, Crise B, Burgess SM (2003) Transcription start regions in the human ge-nome are favored targets for MLV integration. Science 300(5626):1749–1751.

4. Shun M-C, et al. (2007) LEDGF/p75 functions downstream from preintegration complexformation to effect gene-specific HIV-1 integration. Genes Dev 21(14):1767–1778.

5. Mitchell RS, et al. (2004) Retroviral DNA integration: ASLV, HIV, and MLV show dis-tinct target site preferences. PLoS Biol 2(8):E234.

6. Fassati A, Goff SP (1999) Characterization of intracellular reverse transcription com-plexes of Moloney murine leukemia virus. J Virol 73(11):8919–8925.

7. Bowerman B, Brown PO, Bishop JM, Varmus HE (1989) A nucleoprotein complexmediates the integration of retroviral DNA. Genes Dev 3(4):469–478.

8. Prizan-Ravid A, et al. (2010) The Gag cleavage product, p12, is a functional constituentof the murine leukemia virus pre-integration complex. PLoS Pathog 6(11):e1001183.

9. Suzuki Y, Craigie R (2002) Regulatory mechanisms by which barrier-to-autointegrationfactor blocks autointegration and stimulates intermolecular integration of Moloneymurine leukemia virus preintegration complexes. J Virol 76(23):12376–12380.

10. Yuan B, Campbell S, Bacharach E, Rein A, Goff SP (2000) Infectivity of Moloney murineleukemia virus defective in late assembly events is restored by late assembly domainsof other retroviruses. J Virol 74(16):7250–7260.

11. Yuan B, Li X, Goff SP (1999) Mutations altering the Moloney murine leukemia virusp12 Gag protein affect virion production and early events of the virus life cycle. EMBOJ 18(17):4700–4710.

12. Wight DJ, et al. (2012) The gammaretroviral p12 protein has multiple domains thatfunction during the early stages of replication. Retrovirology 9:83.

13. Levin HL, Moran JV (2011) Dynamic interactions between transposable elements andtheir hosts. Nat Rev Genet 12(9):615–627.

14. Engelman A, Cherepanov P (2008) The lentiviral integrase binding protein LEDGF/p75and HIV-1 replication. PLoS Pathog 4(3):e1000046.

15. Llano M, Morrison J, Poeschla EM (2009) Virological and cellular roles of the tran-scriptional coactivator LEDGF/p75. Curr Top Microbiol Immunol 339:125–146.

16. Tobaly-Tapiero J, et al. (2008) Chromatin tethering of incoming foamy virus by thestructural Gag protein. Traffic 9(10):1717–1727.

17. Barbera AJ, et al. (2006) The nucleosomal surface as a docking station for Kaposi’ssarcoma herpesvirus LANA. Science 311(5762):856–861.

18. Sekhar V, McBride AA (2012) Phosphorylation regulates binding of the human pap-illomavirus type 8 E2 protein to host chromosomes. J Virol 86(18):10047–10058.

19. Sekhar V, Reed SC, McBride AA (2010) Interaction of the betapapillomavirus E2tethering protein with mitotic chromosomes. J Virol 84(1):543–557.

20. Voelkel C, et al. (2010) Protein transduction from retroviral Gag precursors. Proc NatlAcad Sci USA 107(17):7805–7810.

21. Yu JH, Schaffer DV (2006) High-throughput, library-based selection of a murineleukemia virus variant to infect nondividing cells. J Virol 80(18):8981–8988.

22. Albritton LM, Tseng L, Scadden D, Cunningham JM (1989) A putative murine eco-

tropic retrovirus receptor gene encodes a multiple membrane-spanning protein and

confers susceptibility to virus infection. Cell 57(4):659–666.23. Goff SP, Traktman P, Baltimore D (1981) Isolation and properties of Moloney murine

leukemia virus mutants: Use of a rapid assay for release of virion reverse transcriptase.

J Virol 38(1):239–248.24. Yueh A, Goff SP (2003) Phosphorylated serine residues and an arginine-rich domain of

the Moloney murine leukemia virus p12 protein are required for early events of viral

infection. J Virol 77(3):1820–1829.25. Elis E, EhrlichM, Prizan-Ravid A, Laham-KaramN, Bacharach E (2012) p12 tethers themurine

leukemia virus pre-integration complex to mitotic chromosomes. PLoS Pathog 8(12):

e1003103.26. Lewinski MK, et al. (2006) Retroviral DNA integration: Viral and cellular determinants

of target-site selection. PLoS Pathog 2(6):e60.27. Cherezova L, Burnside KL, Rose TM (2011) Conservation of complex nuclear locali-

zation signals utilizing classical and non-classical nuclear import pathways in LANA

homologs of KSHV and RFHV. PLoS One 6(4):e18920.28. Weiss MA, Narayana N (1998) RNA recognition by arginine-rich peptide motifs. Bio-

polymers 48(2-3):167–180.29. Shire K, et al. (2006) Regulation of the EBNA1 Epstein-Barr virus protein by serine

phosphorylation and arginine methylation. J Virol 80(11):5261–5272.30. Sen A, Sherr CJ, Todaro GJ (1976) Specific binding of the type C viral core protein p12

with purified viral RNA. Cell 7(1):21–32.31. Felkner RH, Roth MJ (1992) Mutational analysis of the N-linked glycosylation sites of

the SU envelope protein of Moloney murine leukemia virus. J Virol 66(7):4258–4264.32. Chen C-C, Rivera A, Ron N, Dougherty JP, Ron Y (2001) A gene therapy approach for

treating T-cell-mediated autoimmune diseases. Blood 97(4):886–894.33. O’Reilly L, Roth MJ (2000) Second-site changes affect viability of amphotropic/eco-

tropic chimeric enveloped murine leukemia viruses. J Virol 74(2):899–913.34. Cosset FL, Takeuchi Y, Battini JL, Weiss RA, Collins MK (1995) High-titer packaging cells

producing recombinant retroviruses resistant to human serum. J Virol 69(12):7430–7436.35. Fouchier RAM, Meyer BE, Simon JHM, Fischer U, Malim MH (1997) HIV-1 infection of non-

dividing cells: Evidence that the amino-terminal basic region of the viral matrix protein is

important for Gag processing but not for post-entry nuclear import. EMBO J 16(15):

4531–4539.36. Soneoka Y, et al. (1995) A transient three-plasmid expression system for the pro-

duction of high titer retroviral vectors. Nucleic Acids Res 23(4):628–633.37. Trobridge GD, et al. (2006) Foamy virus vector integration sites in normal human cells.

Proc Natl Acad Sci USA 103(5):1498–1503.38. Ciuffi A, et al. (2005) A role for LEDGF/p75 in targeting HIV DNA integration. Nat Med

11(12):1287–1289.39. Brady T, et al. (2009) Integration target site selection by a resurrected human en-

dogenous retrovirus. Genes Dev 23(5):633–642.

9492 | www.pnas.org/cgi/doi/10.1073/pnas.1221736110 Schneider et al.