Skip to main content
  • Submit
  • About
    • Editorial Board
    • PNAS Staff
    • FAQ
    • Accessibility Statement
    • Rights and Permissions
    • Site Map
  • Contact
  • Journal Club
  • Subscribe
    • Subscription Rates
    • Subscriptions FAQ
    • Open Access
    • Recommend PNAS to Your Librarian
  • Log in
  • My Cart

Main menu

  • Home
  • Articles
    • Current
    • Special Feature Articles - Most Recent
    • Special Features
    • Colloquia
    • Collected Articles
    • PNAS Classics
    • List of Issues
  • Front Matter
  • News
    • For the Press
    • This Week In PNAS
    • PNAS in the News
  • Podcasts
  • Authors
    • Information for Authors
    • Editorial and Journal Policies
    • Submission Procedures
    • Fees and Licenses
  • Submit
  • About
    • Editorial Board
    • PNAS Staff
    • FAQ
    • Accessibility Statement
    • Rights and Permissions
    • Site Map
  • Contact
  • Journal Club
  • Subscribe
    • Subscription Rates
    • Subscriptions FAQ
    • Open Access
    • Recommend PNAS to Your Librarian

User menu

  • Log in
  • My Cart

Search

  • Advanced search
Home
Home

Advanced Search

  • Home
  • Articles
    • Current
    • Special Feature Articles - Most Recent
    • Special Features
    • Colloquia
    • Collected Articles
    • PNAS Classics
    • List of Issues
  • Front Matter
  • News
    • For the Press
    • This Week In PNAS
    • PNAS in the News
  • Podcasts
  • Authors
    • Information for Authors
    • Editorial and Journal Policies
    • Submission Procedures
    • Fees and Licenses

New Research In

Physical Sciences

Featured Portals

  • Physics
  • Chemistry
  • Sustainability Science

Articles by Topic

  • Applied Mathematics
  • Applied Physical Sciences
  • Astronomy
  • Computer Sciences
  • Earth, Atmospheric, and Planetary Sciences
  • Engineering
  • Environmental Sciences
  • Mathematics
  • Statistics

Social Sciences

Featured Portals

  • Anthropology
  • Sustainability Science

Articles by Topic

  • Economic Sciences
  • Environmental Sciences
  • Political Sciences
  • Psychological and Cognitive Sciences
  • Social Sciences

Biological Sciences

Featured Portals

  • Sustainability Science

Articles by Topic

  • Agricultural Sciences
  • Anthropology
  • Applied Biological Sciences
  • Biochemistry
  • Biophysics and Computational Biology
  • Cell Biology
  • Developmental Biology
  • Ecology
  • Environmental Sciences
  • Evolution
  • Genetics
  • Immunology and Inflammation
  • Medical Sciences
  • Microbiology
  • Neuroscience
  • Pharmacology
  • Physiology
  • Plant Biology
  • Population Biology
  • Psychological and Cognitive Sciences
  • Sustainability Science
  • Systems Biology
Research Article

Epstein-Barr virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing p16INK4A and p14ARF expression

Seiji Maruo, Bo Zhao, Eric Johannsen, Elliott Kieff, James Zou, and Kenzo Takada
PNAS February 1, 2011 108 (5) 1919-1924; https://doi.org/10.1073/pnas.1019599108
Seiji Maruo
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: ekieff@rics.bwh.harvard.edu smaruo@igm.hokudai.ac.jp
Bo Zhao
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Eric Johannsen
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Elliott Kieff
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: ekieff@rics.bwh.harvard.edu smaruo@igm.hokudai.ac.jp
James Zou
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kenzo Takada
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  1. Contributed by Elliott Kieff, December 29, 2010 (sent for review December 21, 2010)

  • Article
  • Figures & SI
  • Info & Metrics
  • PDF
Loading

Abstract

Epstein-Barr virus (EBV) nuclear antigen 3C (EBNA3C) and EBNA3A are each essential for EBV conversion of primary human B lymphocytes into continuously proliferating lymphoblast cell lines (LCLs) and for maintaining LCL growth. We now find that EBNA3C and EBNA3A's essential roles are to repress p16INK4A and p14ARF. In the absence of EBNA3C or EBNA3A, p16INK4A and p14ARF expression increased and cell growth ceased. EBNA3C inactivation did not alter p16INK4A promoter CpG methylation, but reduced already low H3K27me3, relative to resting B cells, and increased H3K4me3 and H3-acetylation, linking EBNA3C inactivation to histone modifications associated with increased transcription. Importantly, knockdown of p16INK4A or p14ARF partially rescued LCLs from EBNA3C or EBNA3A inactivation-induced growth arrest and knockdown of both rescued LCL growth, confirming central roles for p16INK4A and p14ARF in LCL growth arrest following EBNA3C or EBNA3A inactivation. Moreover, blockade of p16INK4A and p14ARF effects on pRb and p53 by human papilloma virus type 16 E7 and E6 expression, sustained LCL growth after EBNA3C or EBNA3A inactivation. These data indicate that EBNA3C and EBNA3A joint repression of CDKN2A p16INK4A and p14ARF is essential for LCL growth.

  • cell cycle
  • lymphoma
  • Notch
  • CtBP
  • polycomb protein

Epstein-Barr virus (EBV) is causally associated with lymphoproliferative diseases, endemic Burkitt lymphomas, Hodgkin lymphomas, other B- and T-cell lymphomas, anaplastic nasopharyngeal carcinomas, and gastric carcinomas (reviewed in ref. 1). In primary human infection, and in vitro, EBV latency III infection converts B cells into proliferating lymphoblast cell lines (LCLs) (2, 3). In LCLs, EBV expresses six nuclear antigen proteins (EBNA2, EBNALP, EBNA3A, EBNA3B, EBNA3C, and EBNA1), two latent infection membrane proteins (LMP1 and LMP2), two small RNAs (EBER1 and EBER2), and BamHI A rightward transcripts (BARTs). EBNA and BART transcripts also encode multiple micro-RNAs. Reverse genetic and biochemical analyses indicate that EBNA2, EBNALP, EBNA3A, EBNA3C, and LMP1 mediate LCL growth, whereas EBNA1 maintains and enhances transcription from EBV episome, and LMP2 is important for immortalization of less mature B cells. EBNA2 activates and EBNALP coactivates MYC, EBNA3A, EBNA3B, EBNA3C, EBNA1, LMP1, and LMP2 transcription (reviewed in ref. 4).

The experiments described here investigate the underlying biochemical mechanisms and significance of initial findings that conditional EBNA3A or EBNA3C inactivation in LCLs induces G1 growth arrest, which for EBNA3C is associated with induction of CDKN2A p16INK4A expression, consistent with the possibility that EBNA3A and EBNA3C's essential role is to repress CDKN2A p16INK4A (5–8). EBNA3A, EBNA3B, and EBNA3C likely arose as a consequence of gene triplication, after the evolution of EBNA2 and EBNALP (9). EBNA3A, EBNA3B, and EBNA3C have the same promoter, similar exons, codons, introns, and a homologous domain, which strongly interacts with RBPJ, the sequence-specific DNA binding transcription factor that mediates EBNA2 up-regulation of transcription (10–14). Despite these similarities, EBNA3A, EBNA3B, and EBNA3C have divergent roles in maintaining LCL growth, because EBNA3B is dispensable, and only EBNA3A can replace a conditionally inactivated EBNA3A, and only EBNA3C can replace a conditionally inactivated EBNA3C (5, 6, 15–17). Because EBNA2 activates MYC expression through RBPJ, and associates less stably with RBPJ than EBNA3A, EBNA3B, or EBNA3C, some EBNA3 effects on transcription and LCL growth may be in limitation of EBNA2 access to RBPJ (10–14, 18–21). Indeed, all mutations in EBNA3A or EBNA3C that inhibit association with RBPJ are null for LCL growth (22–24), whereas EBNA3A or EBNA3C mutations that affect binding to the adenovirus E1a C-terminal binding protein (CtBP) corepressor result in continuous, albeit slower, growth (22–26). EBNA3C or EBNA3A have many specific and potentially significant interactions with other transcription factors or modifiers, including PU.1, Spi-B, HDAC1, DP103, prothymosin-α, p300, Nm23-H1, SUMO1, and SUMO3, cyclin A, SCFSKP2 ubiquitin ligase, pRb, Chk2, Mdm2, and MRS18-2, and these interactions could be relevant to CDKN2A p16INK4A or p14ARF regulation and LCL growth (27–38). Furthermore, EBNA3C up-regulates TCL1A and ITGA4, down-regulates JAG1 and NCALD RNAs, and cooperates with EBNA3A in repressing Bim, a proapoptotic Bcl-2 family protein (24, 39–41). Indeed, EBNA3C and EBNA3A may cooperatively repress the p16INK4A and p14ARF tumor suppressors to allow cell cycle progression, as is required for MYC conversion of tissue cells to stem cells, for HPV16- and HPV18-induced cervical cancer, and for other enforced cell proliferations (42–44).

Results

EBNA3C Inactivation Induces p16INK4A and p14ARF in LCLs.

LCLs infected with recombinant EBV genomes that express a conditionally active EBNA3C fused to a 4-hydroxytamoxifen (4HT)-dependent mutant estrogen-receptor hormone-binding domain stably express E3CHT in media with 4HT and grow at rates similar to wild-type LCLs (6, 45). In media without 4HT, E3CHT levels substantially decrease by 5–10 d and the LCLs stop growing by day 10, or soon thereafter (Fig. 1A, Fig. S1A, and ref. 6), unless wild-type EBNA3C is expressed, in trans, to maintain LCL growth (Fig. S1B and ref. 6). In media without 4HT, at day 6 and 11, CDKN2A p16INK4A and p14ARF RNAs significantly, substantially, and progressively increased and at day 5, 10, and 15 CDKN2A p16INK4A and p14ARF protein expression also substantially and progressively increased (Fig. 1 A–C and refs. 6 and 8). In contrast, p16INK4A and p14ARF RNA remained low in the E3CHT LCLs cultured with 4HT at day 6 and 11, as did protein levels at day 5, 10, and 15 (Fig. 1 A–C and refs. 6 and 8). Further, wild-type EBNA3C expression from an oriP plasmid transfected into E3CHT LCLs, before transfer to media without 4HT, prevented p16INK4A and p14ARF RNA induction after E3CHT inactivation and sustained LCL growth in the absence of 4HT (Fig. 1D and Fig. S1B). These results indicate that E3CHT inactivation induces p16INK4A and p14ARF RNA and protein expression and LCL growth arrest, whereas wild-type EBNA3C expressed in the same cells prevents p16INK4A and p14ARF RNA and protein expression and maintains LCL growth.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

EBNA3C inactivation results in the induction of p16INK4A and p14ARF in LCLs. (A) Lysates of E3CHT LCLs cultured in the absence (−) or presence (+) of 4HT for the indicated days were Western blotted with EBV-immune human or p16INK4A, p14ARF, or actin antibodies. n.s., nonspecific. (B) Drawing of the CDKN2A locus p16INK4A and p14ARF mRNAs and p16INK4A (dark gray) and p14ARF (light gray) ORFs. Dashes below p16INK4A and p14ARF unique exons indicate shRNA targets and arrows, primers for real-time qRT-PCR. p14ARF exon 1β is upstream of p16INK4A exon 1α. (C) RNA was extracted from E3CHT LCLs in medium with (+) or without (−) 4HT for 6 or 11 d. Real-time p16INK4A and p14ARF qRT-PCR was normalized to GAPDH. Each bar is a three determination-based SEM. (D) E3CHT LCLs transfected with an oriP plasmid expressing wild-type EBNA3C (E3C) or a control oriP plasmid (Cont) and 5 d later (day 0), were washed and transferred to medium with (+) or without (−) 4HT. RNA was extracted at day 29 and real-time qRT-PCR analyzed for p16INK4A and p14ARF levels.

EBNA3C Inactivation Does Not Change p16INK4A and p14ARF Promoter CpG Methylation.

Because the p16INK4A locus is frequently silenced by CpG promoter methylation in human tumors (46), we examined whether E3CHT inactivation decreases CDKN2A locus CpG methylation in LCLs, using methylation-specific PCR (MSP). Methylated CpG-specific amplifications were readily detected at the p16INK4A and p14ARF promoters in Burkitt lymphoma (BL) cell lines (Fig. 2A). In contrast, unmethylated CpG-specific amplifications, but not methylated CpG-specific amplifications, were detected at the p16INK4A and p14ARF promoters in E3CHT LCLs, in the absence or presence of 4HT (Fig. 2A). Using bisulfite sequencing, CpGs in the p16INK4A promoter were almost fully methylated in BL cells, whereas most CpGs were unmethylated in E3CHT LCLs in the absence or presence of E3CHT (Fig. 2B), consistent with the methylation-specific PCR results (Fig. 2A). Thus, EBNA3C represses CDKN2A p16INK4A and p14ARF expression without significantly altering the mostly nonmethylated CpG state in LCLs.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

EBNA3C inactivation does not alter p14ARF or p16INK4A promoter CpG methylation in LCLs. (A) Akata Burkitt lymphoma (BL), P3HR-1 BL, and E3CHT LCL DNAs cultured in the absence (−) or presence (+) of 4HT for 41 d were subjected to methylation-specific PCR using p16INK4A- or p14ARF-specific primers for the methylated (M) or unmethylated (U) promoter sequences. (B) The methylation state of CpGs in the p16INK4A promoter in P3HR-1 BL and E3CHT LCLs cultured in the absence or presence of 4HT for 41 d was determined by bisulfite sequencing.

EBNA3C Inactivation Changes Chromatin at the p16INK4A Locus from Repressive to Active.

High-level histone H3K27 trimethylation (H3K27me3) and low level H3K4me3 are associated with p16INK4A silencing (47). ChIP analyses indicated that E3CHT inactivation significantly decreased H3K27me3 and increased H3K4me3 within 0.6 kb upstream and 0.5 kb downstream of the p16INK4A promoter in E3CHT LCLs (Fig. 3), indicative of a shift from repressive to active chromatin. In further support of a more active chromatin state, E3CHT inactivation increased H3 acetylation at the same sites (Fig. 3, AcH3).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

EBNA3C inactivation increases p16INK4A histone H3 acetylation and H3K4me3 and reduces of H3K27 trimethylation in LCLs. E3CHT LCLs cultured in the absence (−) or presence (+) of 4HT for 20 d were ChIP and qRT-PCR analyzed with the indicated antibodies and primer sets are shown and listed in Table S1. Bars indicate the mean and SE of three reactions.

Comparison of active transcription-associated H3K4me3 and H3K36me3 histone modifications with Polycomb repression-associated H3K27me3 marks at the CDKN2A p16INK4A and p14ARF in LCLs relative to primary CD19 positive B cells using normalized Encyclopedia of DNA Elements (ENCODE) data (Fig. S2) revealed extensive H3K27me3 through the CDKN2A p16INK4A and p14ARF locus in resting CD19+ primary B cells, except at the p16INK4A and p14ARF promoters versus very little H3K27me3 throughout the locus in LCLs. Whereas H3K36me3 was low through the locus in both resting B cells and LCLs, H3K4me3 levels were high at the p16INK4A and p14ARF promoters. The coupling of low overall H3K27me3 and high H3K4me3 at the p16INK4A and p14ARF promoters, with above background PolII through these loci in LCLs (Fig. S2) is consistent with p16INK4A and p14ARF being poised for activation in LCLs.

p16INK4A and p14ARF Knockdown Rescues LCLs from Growth Arrest Following EBNA3C Inactivation.

To evaluate whether p16INK4A and p14ARF are responsible for the LCL growth arrest induced by EBNA3C inactivation, oriP plasmids that express shRNAs targeting p16INK4A or p14ARF were verified to knock down their target RNAs in 293T cells by Western blot for p16INK4A and p14ARF (Fig. S3). When E3CHT LCLs were transfected with oriP plasmids that express shRNA targeting p16INK4A or p14ARF or p16INK4A and p14ARF, and transfected LCLs were cultured in medium with or without 4HT, knockdown of p16INK4A or p14ARF partially sustained E3CHT LCL growth in medium without 4HT and knockdown of p16INK4A and p14ARF fully rescued LCL growth (Fig. 4A). Western blot confirmed the knockdown of p16INK4A and p14ARF proteins in E3CHT LCLs following transfection with both p16INK4A and p14ARF shRNAs and culture in medium without 4HT (Fig. 4B). These data indicate that EBNA3C suppression of p16INK4A and p14ARF is the principal basis for EBNA3C's essential role in LCL growth.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Knockdown of p16INK4A and p14ARF rescues LCLs from EBNA3C inactivation-induced growth arrest. (A) E3CHT LCLs were transfected with an oriP plasmid expressing EBNA3C (E3C) or an oriP plasmid expressing control shRNA (sh-cont), shRNA targeting p16INK4A (sh-p16), shRNA targeting p14ARF (sh-p14), or both sh-p16– and sh-p14–targeting plasmids. Five days after transfection (day 0), cells were washed and transferred to medium with (+) or without (−) 4HT, and cell growth was determined. (B) Total cell lysates were prepared in A at day 41 and Western blot analyzed with EBV-immune human serum, p16INK4A-, p14ARF-, or actin-specific antibodies. Arrowhead indicates transfected E3C and n.s. indicates nonspecific. (C) E3CHT LCLs were transfected with an oriP plasmid expressing EBNA3C (E3C), HPV16 E7, HPV16 E6, both oriP plasmids expressing E7 and E6 (E7 + E6), or a control oriP plasmid (Cont). Cell growth was determined in medium with (+) or without (−) 4HT.

Because human papilloma virus type 16 (HPV16) E7 and E6 inactivate pRb and p53, downstream targets of p16INK4A and p14ARF, respectively, in cell growth inhibition (43, 44), E6 and E7 were used to confirm the key role for p16INK4A and p14ARF and to investigate the importance of pRB and p53 inactivation in EBNA3C effects. Expression of either E7 or E6 in E3CHT LCLs partially rescued LCLs from growth arrest after E3CHT inactivation, whereas E7 and E6 expression fully rescued LCL growth (Fig. 4C). E7 and E6 rescue of LCL growth was similar to that of shRNAs targeting p16INK4A and p14ARF (Fig. 4 A and C), except that pRb was further decreased and p16INK4A and p14ARF were further increased (Fig. S4). Thus, these data indicate that EBNA3C repression of p16INK4A and p14ARF or their effects on pRb and p53 is essential for LCL growth and implicate pRb and p53 in p16INK4A and p14ARF effects.

EBNA3A Is also Required for p16INK4A and p14ARF Repression.

Because conditional E3AHT inactivation in LCLs has similar effects on LCL growth and p16INK4A is also elevated in EBNA3A knockout LCLs relative to wild-type LCLs (5, 7, 8), EBNA3A may also have a role in p16INK4A and p14ARF repression. Indeed, incubation of E3AHT LCLs in media without 4HT, resulted at day 6 in substantially reduced E3AHT, slowed cell growth, increased p16INK4A and p14ARF protein expression, and ∼1.4-fold significantly increased p16INK4A and p14ARF RNA levels (Fig. 5 A and B). E3AHT LCL growth stopped by day 10 and at day 12, p16INK4A and p14ARF RNA were 2- and 1.7-fold increased and p16INK4A and p14ARF proteins were at very high levels (Fig. 5). Importantly, shRNA knockdown of p16INK4A and p14ARF restored EBNA3A-dependent LCL growth (Fig. 5C). Further, HPV16 E7 and E6 expression similarly sustained LCLs following E3AHT inactivation (Fig. 5D), confirming the importance of EBNA3A repression of the p16INK4A and p14ARF pathways for continued LCL growth.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

EBNA3A inactivation induces p16INK4A and p14ARF expression and LCL growth arrest. (A) Lysates of E3AHT LCLs, cultured in the absence (−) or presence (+) of 4HT, for the indicated days, were Western blotted with EBV-immune human serum or p16INK4A-, p14ARF-, or actin-specific antibodies. n.s., nonspecific. (B) RNA was extracted from E3AHT LCLs in medium with (+) or without (−) 4HT for the indicated days. Real-time qRT-PCRs for p16INK4A and p14ARF RNA levels were normalized against GAPDH. Each bar is the mean of three reactions and the SE. (C) E3AHT LCLs were transfected with an oriP plasmid expressing EBNA3A (E3A), control shRNA (sh-cont), shRNA targeting p16INK4A (sh-p16), shRNA targeting p14ARF (sh-p14), both oriP plasmids expressing sh-p16 and sh-p14 (sh-p16 + sh-p14), or a negative control oriP plasmid (Cont). Seven days after transfection (day 0), cells were washed and transferred to medium with (+) or without (−) 4HT, and subsequent cell growth was determined. (D) E3AHT LCLs were transfected with an oriP plasmid expressing EBNA3A (E3A), HPV16 E7, HPV16 E6, both oriP plasmids expressing E7 and E6 (E7 + E6), or a control oriP plasmid (Cont). Cell growth was determined in medium with (+) or without (−) 4HT.

Discussion

The data presented here demonstrate that the E3CHT or E3AHT inactivation in LCLs induces CDKN2A p16INK4A and p14ARF expression and that knockdown of p16INK4A or p14ARF partially sustains LCL growth, whereas knockdown of both p16INK4A and p14ARF maintains LCL growth. These data, together with previous data that EBNA3A and EBNA3C are each essential for LCL growth (5, 6) and cannot be replaced by increased expression of the other, indicate that EBNA3A and EBNA3C have cooperative and independent essential roles in maintaining LCL growth by repressing CDKN2A p16INK4A and p14ARFexpression.

These effects on LCL growth are likely at least partially mediated by p16INK4A effects on pRb stability because pRb levels fall as EBNA3C inactivation induces p16INK4A and p14ARF expression (Fig. S4). Consistent with an important role for p16INK4A effects on pRB and of p14ARF on p53, HPV E7 or E6 partially maintained LCL growth following EBNA3C or EBNA3A inactivation and HPV E7 and E6 restored LCL growth.

The mechanisms through which EBNA3A and EBNA3C suppress CDKN2A p16INK4A and p14ARF are incompletely understood, but may be determined by the fundamental mechanisms through which CDKN2A p16INK4A and p14ARF are induced and regulated in latency III EBV infection. EBNA2 is expressed first and strongly up-regulates MYC (48–50). MYC induces cyclin D2, and cyclin D2-enforced cell cycle entry likely induces CDKN2A p16 and p14 expression (48–52). CDKN2A p16INK4A and p14ARF are partially regulated by different promoters and have different first exons (exon 1α for p16INK4A and 1β for p14ARF), but share exons 2 and 3 (53, 54). The CDKN2A p16INK4A-pRb and the p14ARF-p53 pathways are inactivated in many cancers and by DNA viruses, which force infected cells into S phase to enable virus DNA replication. High-risk HPV E7 and E6 oncoproteins disrupt the p16INK4A-pRb and p14ARF-p53 pathways by targeting downstream components, pRB and p53 (55). EBNA proteins are also expressed in virus replication and may induce MYC to support virus replication (56). Nevertheless, EBNA2 induction of MYC and EBNA3A and EBNA3C suppression of CDKN2A p16INK4A and p14ARF expression are essential for latency III EBV infected cell proliferation, which enables long-term latency in human lymphoid organs (for review, see ref. 1).

This study indicates that EBNA3C and EBNA3A's principal essential role in LCL growth is to suppress cell proliferation–induced activation of the CDKN2A p16INK4A and p14ARF pathways to prevent p16INK4A and p14ARF inhibition of cell growth. Although E7 and E6 expression rescued LCL cell growth, E6 and E7 further increased p16INK4A and p14ARF expression (Fig. S4), as described with HPV infection (57), probably as a consequence of E7-induced Rb degradation (Fig. S4 and ref. 58).

DNA methylation was not evident at the CDKN2A p16INK4A and p14ARF promoters in E3CHT on or off LCLs, indicating that DNA methylation does not have a major role in EBNA3C repression of this locus. Although Polycomb group (PcG) protein-associated H3K27me3 modifications and JMJD3 demethylase are implicated in CDKN2A locus regulation (54, 59–61), the ENCODE LCL data shown in Fig. S2 indicate that the CDKN2A p16INK4A and p14ARF locus in LCLs is already deficient in H3K27me3 and has over-background Pol II, as well as promoter-associated H3K4me3, consistent with significant potential for further derepression and activation. Indeed, EBNA3C inactivation further reduced H3K27me3 at the p16INK4A promoter and increased histone H3 acetylation and H3K4me3 consistent with recent findings of EBNA3C- and EBNA3A-related histone modification at the p16INK4A locus (8).

Knockdown of p14ARF was also required for restoration of LCL growth in EBNA3C- or EBNA3A-inactivated LCLs, indicating the significance of EBNA3C and EBNA3A p14ARF suppression. Interestingly, p14ARF protein was near or below detection levels in EBNA3A− LCLs (7), which required months on feeder layers for long-term outgrowth. Perhaps selection for p14ARF suppression occurred during the initial growth on fibroblast feeder cells. The EBNA3A− LCLs continuously grew with reduced proliferation rates, perhaps due to p16INK4A induction.

The mechanisms by which EBNA3C and EBNA3A repress CDKN2A p16INK4A and p14ARF expression remain to be elucidated. Certainly, constitutive EBNA2-mediated c-myc up-regulation has a central role in EBV-mediated LCL growth (49, 62) and almost certainly underlies p16INK4A and p14ARF induction in EBV-infected B cells in the absence of EBNA3C or EBNA3A. Forced MYC expression induces p16INK4A and p14ARF in normal human diploid fibroblasts and megakaryocytes (42, 63). Alternatively, aberrant cell cycle progression induced by EBNA2 may induce p16INK4A and p14ARF through E2F or CDC6 (50, 53, 64, 65). EBNA3C and EBNA3A association with RBPJ may competitively modulate EBNA2 effects through RBPJ on MYC up-regulation so that EBV-infected B cells proliferate at a lower rate or with a compensatory EBNA3C or EBNA3A RBPJ-mediated effect on CDKN2A p16INK4A and p14ARF suppression.

Because only EBNA3C can transcomplement a conditionally inactivated EBNA3C and only EBNA3A can transcomplement a conditionally inactivated EBNA3A, both EBNA3C and EBNA3A are necessary for effective p16INK4A and p14ARF repression. Also, EBNA3B, which interacts with RBPJ is not required for LCL proliferation (17). EBNA3C and EBNA3A association with CtBP has been implicated in epigenetic repression of p16INK4A (8), although deletion of the EBNA3C or EBNA3A binding sites for CtBP is only associated with slower LCL growth, whereas even point mutations in the EBNA3C or EBNA3A binding sites for RBPJ are null for LCL growth (22–24). EBNA3C and/or EBNA3A may cooperatively modulate transcription of regulators of the CDKN2A locus or more directly regulate suppression of p16INK4A and p14ARF through another interaction with RBPJ.

Materials and Methods

Cell Lines and Plasmids.

E3CHT and E3AHT LCLs are infected with a recombinant EBV, which express 4-hydroxy-tamoxifen (4HT)-dependent EBNA3C (E3CHT) or EBNA3A (E3AHT) (5, 6). In some experiments, E3CHT or E3AHT LCLs were transcomplemented with EBNA3A or EBNA3C expressed from OriP plasmids (5, 6). HPV16 E6 or E7 (65) were kindly provided by T. Kiyono (National Cancer Center Research Institute, Tokyo, Japan) and cloned into an oriP plasmid. The oriP plasmids expressing shRNA have a hygromycin resistance cassette derived from pSilencer 2.1-U6 hygro (Ambion) and a human U6 RNA pol III promoter-driving shRNA targeting p16INK4A, p14ARF, or control shRNA (Ambion), subcloned into an XbaI–NruI-digested pCEP4 vector (Invitrogen). The shRNA-targeted sequences were as follows: p16INK4A, 5′-TGCCCAACGCACCGAATAGTTACGGTC-3′ and p14ARF, 5′-GAACATGGTGCGCAGGTTC-3′.

Western Blot Analysis.

Western blot analyses (6) used anti-p16 (Santa Cruz), anti-p15 (Santa Cruz), anti-p14 (Lab Vision), anti-Rb (PharMingen), and anti–β-actin (Sigma) antibodies.

Quantitative RT-PCR.

Total RNA extraction, cDNA synthesis, and real-time qRT-PCR (6) used primers for amplification of specific targets including p16 forward (5′-AGCATGGAGCCTTCGGCTGA-3′, located on exon 1α) and reverse (5′-CCATCATCATGACCTGGATCG-3′, located on the junction of exon 1α and exon 2); p14 forward (5′-TACTGAGGAGCCAGCGTCTA-3′, located on exon 1β) and reverse (5′-TGCACGGGTCGGGTGAGAGT-3′, located on exon 2); and GAPDH forward (5′-GCCTCCTGCACCACCAACTG-3′) and reverse (5′-CGACGCCTGCTTCACCACCTTCT-3′).

MSP Analysis.

Genome DNA was subjected to bisulfite modification using an EpiTect Bisulfite kit (Qiagen). Modified DNA template was used to amplify the p16INK4A and p14ARF promoters with methylated- or unmethylated-specific primer pairs (66, 67).

Bisulfite Sequencing of p16INK4A Promoter.

Bisulfite-converted DNA was used to amplify the p16INK4A promoter (68). PCR products were cloned using pGEM-T Easy vector system (Promega). Colonies were picked and sequenced.

ChIP Assays.

ChIPs used EZ-ChIP kit (Millipore) and ChIP Ab+ acetyl-histone H3 (17–615; Millipore), ChIP Ab+ trimethyl-histone H3 (Lys27) (17–622; Millipore), and ChIP Ab+ trimethyl-histone H3 (Lys4) (17–614; Millipore). Immunoprecipitated DNAs were quantified by real-time qPCR using a SYBR Premix Ex Taq II kit (Takara) and a LightCycler (Roche) and were normalized to input DNA. Primers are listed in Table S1 (69).

Transcomplementation Assay.

Five to 10 million E3CHT LCLs or E3AHT LCLs were electroporated with 20–30 μg of oriP plasmid DNA. Transfected LCLs were cultured in medium with 4HT for 5–7 d, washed, and resuspended at 5–10 × 105 cells/5 mL of complete medium with or without 4HT in a 25-cm2 culture flask. Every 5–8 d, viable cells were counted using a hemocytometer and trypan blue exclusion and cultures were split. Total viable cells were calculated relative to initial cultures.

Acknowledgments

We thank M. Noguchi for suggestions for ChIP analyses. We also acknowledge Brad Bernstein and the National Human Genome Research Institute ENCODE Chromatin Group at Massachusetts General Hospital/Broad Institute, and the National Institutes of Health Roadmap for Epigenomics Mapping Center at the Broad Institute for the ENCODE data. This work was supported by a Grant-in-Aid for Scientific Research from the Japanese Ministry of Education, Culture, Sports, Science, and Technology (to S.M. and K.T.) and by National Cancer Institute Grant CA47006 (to E.K.).

Footnotes

  • 1To whom correspondence may be addressed. E-mail: ekieff{at}rics.bwh.harvard.edu or smaruo{at}igm.hokudai.ac.jp.
  • Author contributions: S.M., E.K., and K.T. designed research; S.M. performed research; B.Z., E.J., J.Z., and K.T. contributed new reagents/analytic tools; S.M. and E.K. analyzed data; and S.M. and E.K. wrote the paper.

  • The authors declare no conflict of interest.

  • This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1019599108/-/DCSupplemental.

View Abstract

References

  1. ↵
    1. Knipe DM,
    2. Howley PM
    1. Rickinson AB,
    2. Kieff ED
    (2007) in Fields Virology, Epstein-Barr virus, eds Knipe DM, Howley PM (Lippincott, Williams and Wilkins, Philadelphia), 2, pp 2655–2700.
    OpenUrl
  2. ↵
    1. Henle W,
    2. Diehl V,
    3. Kohn G,
    4. Zur Hausen H,
    5. Henle G
    (1967) Herpes-type virus and chromosome marker in normal leukocytes after growth with irradiated Burkitt cells. Science 157:1064–1065.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Pope JH
    (1967) Establishment of cell lines from peripheral leucocytes in infectious mononucleosis. Nature 216:810–811.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Knipe DM,
    2. Howley PM
    1. Kieff ED,
    2. Rickinson AB
    (2007) in Fields Virology, Epstein-Barr virus and its replication, eds Knipe DM, Howley PM (Lippincott, Williams and Wilkins, Philadelphia), 2, pp 2603–2654.
    OpenUrl
  5. ↵
    1. Maruo S,
    2. Johannsen E,
    3. Illanes D,
    4. Cooper A,
    5. Kieff E
    (2003) Epstein-Barr virus nuclear protein EBNA3A is critical for maintaining lymphoblastoid cell line growth. J Virol 77:10437–10447.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Maruo S,
    2. et al.
    (2006) Epstein-Barr virus nuclear protein EBNA3C is required for cell cycle progression and growth maintenance of lymphoblastoid cells. Proc Natl Acad Sci USA 103:19500–19505.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Hertle ML,
    2. et al.
    (2009) Differential gene expression patterns of EBV infected EBNA-3A positive and negative human B lymphocytes. PLoS Pathog 5:e1000506.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Skalska L,
    2. White RE,
    3. Franz M,
    4. Ruhmann M,
    5. Allday MJ
    (2010) Epigenetic repression of p16(INK4A) by latent Epstein-Barr virus requires the interaction of EBNA3A and EBNA3C with CtBP. PLoS Pathog 6:e1000951.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Rivailler P,
    2. Cho YG,
    3. Wang F
    (2002) Complete genomic sequence of an Epstein-Barr virus-related herpesvirus naturally infecting a new world primate: A defining point in the evolution of oncogenic lymphocryptoviruses. J Virol 76:12055–12068.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Johannsen E,
    2. Miller CL,
    3. Grossman SR,
    4. Kieff E
    (1996) EBNA-2 and EBNA-3C extensively and mutually exclusively associate with RBPJkappa in Epstein-Barr virus-transformed B lymphocytes. J Virol 70:4179–4183.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Zhao B,
    2. Marshall DR,
    3. Sample CE
    (1996) A conserved domain of the Epstein-Barr virus nuclear antigens 3A and 3C binds to a discrete domain of Jkappa. J Virol 70:4228–4236.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Robertson ES,
    2. et al.
    (1995) Epstein-Barr virus nuclear protein 3C modulates transcription through interaction with the sequence-specific DNA-binding protein J kappa. J Virol 69:3108–3116.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Robertson ES,
    2. Lin J,
    3. Kieff E
    (1996) The amino-terminal domains of Epstein-Barr virus nuclear proteins 3A, 3B, and 3C interact with RBPJ(kappa) J Virol 70:3068–3074.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Waltzer L,
    2. Perricaudet M,
    3. Sergeant A,
    4. Manet E
    (1996) Epstein-Barr virus EBNA3A and EBNA3C proteins both repress RBP-J kappa-EBNA2-activated transcription by inhibiting the binding of RBP-J kappa to DNA. J Virol 70:5909–5915.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Tomkinson B,
    2. Kieff E
    (1992) Use of second-site homologous recombination to demonstrate that Epstein-Barr virus nuclear protein 3B is not important for lymphocyte infection or growth transformation in vitro. J Virol 66:2893–2903.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Tomkinson B,
    2. Robertson E,
    3. Kieff E
    (1993) Epstein-Barr virus nuclear proteins EBNA-3A and EBNA-3C are essential for B-lymphocyte growth transformation. J Virol 67:2014–2025.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Chen A,
    2. Divisconte M,
    3. Jiang X,
    4. Quink C,
    5. Wang F
    (2005) Epstein-Barr virus with the latent infection nuclear antigen 3B completely deleted is still competent for B-cell growth transformation in vitro. J Virol 79:4506–4509.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Grossman SR,
    2. Johannsen E,
    3. Tong X,
    4. Yalamanchili R,
    5. Kieff E
    (1994) The Epstein-Barr virus nuclear antigen 2 transactivator is directed to response elements by the J kappa recombination signal binding protein. Proc Natl Acad Sci USA 91:7568–7572.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Henkel T,
    2. Ling PD,
    3. Hayward SD,
    4. Peterson MG
    (1994) Mediation of Epstein-Barr virus EBNA2 transactivation by recombination signal-binding protein J kappa. Science 265:92–95.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Cooper A,
    2. et al.
    (2003) EBNA3A association with RBP-Jkappa down-regulates c-myc and Epstein-Barr virus-transformed lymphoblast growth. J Virol 77:999–1010.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Radkov SA,
    2. et al.
    (1997) Epstein-Barr virus EBNA3C represses Cp, the major promoter for EBNA expression, but has no effect on the promoter of the cell gene CD21. J Virol 71:8552–8562.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Maruo S,
    2. et al.
    (2005) Epstein-Barr virus nuclear protein 3A domains essential for growth of lymphoblasts: Transcriptional regulation through RBP-Jkappa/CBF1 is critical. J Virol 79:10171–10179.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Maruo S,
    2. et al.
    (2009) Epstein-Barr virus nuclear protein EBNA3C residues critical for maintaining lymphoblastoid cell growth. Proc Natl Acad Sci USA 106:4419–4424.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Lee S,
    2. et al.
    (2009) Epstein-Barr virus nuclear protein 3C domains necessary for lymphoblastoid cell growth: Interaction with RBP-Jkappa regulates TCL1. J Virol 83:12368–12377.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Hickabottom M,
    2. Parker GA,
    3. Freemont P,
    4. Crook T,
    5. Allday MJ
    (2002) Two nonconsensus sites in the Epstein-Barr virus oncoprotein EBNA3A cooperate to bind the co-repressor carboxyl-terminal-binding protein (CtBP) J Biol Chem 277:47197–47204.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Touitou R,
    2. Hickabottom M,
    3. Parker G,
    4. Crook T,
    5. Allday MJ
    (2001) Physical and functional interactions between the corepressor CtBP and the Epstein-Barr virus nuclear antigen EBNA3C. J Virol 75:7749–7755.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Lin J,
    2. Johannsen E,
    3. Robertson E,
    4. Kieff E
    (2002) Epstein-Barr virus nuclear antigen 3C putative repression domain mediates coactivation of the LMP1 promoter with EBNA-2. J Virol 76:232–242.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Zhao B,
    2. Sample CE
    (2000) Epstein-barr virus nuclear antigen 3C activates the latent membrane protein 1 promoter in the presence of Epstein-Barr virus nuclear antigen 2 through sequences encompassing an spi-1/Spi-B binding site. J Virol 74:5151–5160.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. Cotter MA 2nd.,
    2. Robertson ES
    (2000) Modulation of histone acetyltransferase activity through interaction of epstein-barr nuclear antigen 3C with prothymosin alpha. Mol Cell Biol 20:5722–5735.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Grundhoff AT,
    2. et al.
    (1999) Characterization of DP103, a novel DEAD box protein that binds to the Epstein-Barr virus nuclear proteins EBNA2 and EBNA3C. J Biol Chem 274:19136–19144.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Knight JS,
    2. Lan K,
    3. Subramanian C,
    4. Robertson ES
    (2003) Epstein-Barr virus nuclear antigen 3C recruits histone deacetylase activity and associates with the corepressors mSin3A and NCoR in human B-cell lines. J Virol 77:4261–4272.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Knight JS,
    2. Robertson ES
    (2004) Epstein-Barr virus nuclear antigen 3C regulates cyclin A/p27 complexes and enhances cyclin A-dependent kinase activity. J Virol 78:1981–1991.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    1. Knight JS,
    2. Sharma N,
    3. Robertson ES
    (2005) SCFSkp2 complex targeted by Epstein-Barr virus essential nuclear antigen. Mol Cell Biol 25:1749–1763.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    1. Radkov SA,
    2. et al.
    (1999) Epstein-Barr virus nuclear antigen 3C interacts with histone deacetylase to repress transcription. J Virol 73:5688–5697.
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Subramanian C,
    2. et al.
    (2002) Epstein-Barr virus nuclear antigen 3C and prothymosin alpha interact with the p300 transcriptional coactivator at the CH1 and CH3/HAT domains and cooperate in regulation of transcription and histone acetylation. J Virol 76:4699–4708.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Subramanian C,
    2. Robertson ES
    (2002) The metastatic suppressor Nm23-H1 interacts with EBNA3C at sequences located between the glutamine- and proline-rich domains and can cooperate in activation of transcription. J Virol 76:8702–8709.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    1. Knight JS,
    2. Sharma N,
    3. Kalman DE,
    4. Robertson ES
    (2004) A cyclin-binding motif within the amino-terminal homology domain of EBNA3C binds cyclin A and modulates cyclin A-dependent kinase activity in Epstein-Barr virus-infected cells. J Virol 78:12857–12867.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Knight JS,
    2. Sharma N,
    3. Robertson ES
    (2005) Epstein-Barr virus latent antigen 3C can mediate the degradation of the retinoblastoma protein through an SCF cellular ubiquitin ligase. Proc Natl Acad Sci USA 102:18562–18566.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    1. Anderton E,
    2. et al.
    (2008) Two Epstein-Barr virus (EBV) oncoproteins cooperate to repress expression of the proapoptotic tumour-suppressor Bim: Clues to the pathogenesis of Burkitt's lymphoma. Oncogene 27:421–433.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Paschos K,
    2. et al.
    (2009) Epstein-Barr virus latency in B cells leads to epigenetic repression and CpG methylation of the tumour suppressor gene Bim. PLoS Pathog 5:e1000492.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Chen A,
    2. Zhao B,
    3. Kieff E,
    4. Aster JC,
    5. Wang F
    (2006) EBNA-3B- and EBNA-3C-regulated cellular genes in Epstein-Barr virus-immortalized lymphoblastoid cell lines. J Virol 80:10139–10150.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    1. Takayama N,
    2. et al.
    (2010) Transient activation of c-MYC expression is critical for efficient platelet generation from human induced pluripotent stem cells. J Exp Med 207:2817–2830.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    1. Dyson N,
    2. Howley PM,
    3. Münger K,
    4. Harlow E
    (1989) The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 243:934–937.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    1. Werness BA,
    2. Levine AJ,
    3. Howley PM
    (1990) Association of human papillomavirus types 16 and 18 E6 proteins with p53. Science 248:76–79.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Littlewood TD,
    2. Hancock DC,
    3. Danielian PS,
    4. Parker MG,
    5. Evan GI
    (1995) A modified oestrogen receptor ligand-binding domain as an improved switch for the regulation of heterologous proteins. Nucleic Acids Res 23:1686–1690.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Baylin SB,
    2. Herman JG
    (2000) DNA hypermethylation in tumorigenesis: Epigenetics joins genetics. Trends Genet 16:168–174.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Bracken AP,
    2. Helin K
    (2009) Polycomb group proteins: Navigators of lineage pathways led astray in cancer. Nat Rev Cancer 9:773–784.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Alfieri C,
    2. Birkenbach M,
    3. Kieff E
    (1991) Early events in Epstein-Barr virus infection of human B lymphocytes. Virology 181:595–608.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Nikitin PA,
    2. et al.
    (2010) An ATM/Chk2-mediated DNA damage-responsive signaling pathway suppresses Epstein-Barr virus transformation of primary human B cells. Cell Host Microbe 8:510–522.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Kaiser C,
    2. et al.
    (1999) The proto-oncogene c-myc is a direct target gene of Epstein-Barr virus nuclear antigen 2. J Virol 73:4481–4484.
    OpenUrlAbstract/FREE Full Text
  51. ↵
    1. Spender LC,
    2. et al.
    (1999) Control of cell cycle entry and apoptosis in B lymphocytes infected by Epstein-Barr virus. J Virol 73:4678–4688.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    1. Rui L,
    2. Goodnow CC
    (2006) Lymphoma and the control of B cell growth and differentiation. Curr Mol Med 6:291–308.
    OpenUrlCrossRefPubMed
  53. ↵
    1. Kim WY,
    2. Sharpless NE
    (2006) The regulation of INK4/ARF in cancer and aging. Cell 127:265–275.
    OpenUrlCrossRefPubMed
  54. ↵
    1. Gil J,
    2. Peters G
    (2006) Regulation of the INK4b-ARF-INK4a tumour suppressor locus: All for one or one for all. Nat Rev Mol Cell Biol 7:667–677.
    OpenUrlCrossRefPubMed
  55. ↵
    1. Sherr CJ,
    2. McCormick F
    (2002) The RB and p53 pathways in cancer. Cancer Cell 2:103–112.
    OpenUrlCrossRefPubMed
  56. ↵
    1. Yuan J,
    2. Cahir-McFarland E,
    3. Zhao B,
    4. Kieff E
    (2006) Virus and cell RNAs expressed during Epstein-Barr virus replication. J Virol 80:2548–2565.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    1. Sano T,
    2. Masuda N,
    3. Oyama T,
    4. Nakajima T
    (2002) Overexpression of p16 and p14ARF is associated with human papillomavirus infection in cervical squamous cell carcinoma and dysplasia. Pathol Int 52:375–383.
    OpenUrlCrossRefPubMed
  58. ↵
    1. Giarrè M,
    2. et al.
    (2001) Induction of pRb degradation by the human papillomavirus type 16 E7 protein is essential to efficiently overcome p16INK4a-imposed G1 cell cycle Arrest. J Virol 75:4705–4712.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    1. Simon JA,
    2. Kingston RE
    (2009) Mechanisms of polycomb gene silencing: Knowns and unknowns. Nat Rev Mol Cell Biol 10:697–708.
    OpenUrlPubMed
  60. ↵
    1. Barradas M,
    2. et al.
    (2009) Histone demethylase JMJD3 contributes to epigenetic control of INK4a/ARF by oncogenic RAS. Genes Dev 23:1177–1182.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Agger K,
    2. et al.
    (2009) The H3K27me3 demethylase JMJD3 contributes to the activation of the INK4A-ARF locus in response to oncogene- and stress-induced senescence. Genes Dev 23:1171–1176.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Zhao B,
    2. et al.
    (2006) RNAs induced by Epstein-Barr virus nuclear antigen 2 in lymphoblastoid cell lines. Proc Natl Acad Sci USA 103:1900–1905.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Drayton S,
    2. et al.
    (2003) Tumor suppressor p16INK4a determines sensitivity of human cells to transformation by cooperating cellular oncogenes. Cancer Cell 4:301–310.
    OpenUrlCrossRefPubMed
  64. ↵
    1. Gonzalez S,
    2. et al.
    (2006) Oncogenic activity of Cdc6 through repression of the INK4/ARF locus. Nature 440:702–706.
    OpenUrlCrossRefPubMed
  65. ↵
    1. Kiyono T,
    2. et al.
    (1998) Both Rb/p16INK4a inactivation and telomerase activity are required to immortalize human epithelial cells. Nature 396:84–88.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Herman JG,
    2. Graff JR,
    3. Myöhänen S,
    4. Nelkin BD,
    5. Baylin SB
    (1996) Methylation-specific PCR: A novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 93:9821–9826.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Esteller M,
    2. et al.
    (2000) Hypermethylation-associated inactivation of p14(ARF) is independent of p16(INK4a) methylation and p53 mutational status. Cancer Res 60:129–133.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    1. Ivanova TA,
    2. et al.
    (2007) Up-regulation of expression and lack of 5′ CpG island hypermethylation of p16 INK4a in HPV-positive cervical carcinomas. BMC Cancer 7:47.
    OpenUrlCrossRefPubMed
  69. ↵
    1. Kia SK,
    2. Gorski MM,
    3. Giannakopoulos S,
    4. Verrijzer CP
    (2008) SWI/SNF mediates polycomb eviction and epigenetic reprogramming of the INK4B-ARF-INK4A locus. Mol Cell Biol 28:3457–3464.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Article Alerts
Email Article

Thank you for your interest in spreading the word on PNAS.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Epstein-Barr virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing p16INK4A and p14ARF expression
(Your Name) has sent you a message from PNAS
(Your Name) thought you would like to see the PNAS web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Epstein-Barr virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing p16INK4A and p14ARF expression
Seiji Maruo, Bo Zhao, Eric Johannsen, Elliott Kieff, James Zou, Kenzo Takada
Proceedings of the National Academy of Sciences Feb 2011, 108 (5) 1919-1924; DOI: 10.1073/pnas.1019599108

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Epstein-Barr virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing p16INK4A and p14ARF expression
Seiji Maruo, Bo Zhao, Eric Johannsen, Elliott Kieff, James Zou, Kenzo Takada
Proceedings of the National Academy of Sciences Feb 2011, 108 (5) 1919-1924; DOI: 10.1073/pnas.1019599108
Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Mendeley logo Mendeley
Proceedings of the National Academy of Sciences: 108 (5)
Table of Contents

Submit

Sign up for Article Alerts

Article Classifications

  • Biological Sciences
  • Cell Biology

Jump to section

  • Article
    • Abstract
    • Results
    • Discussion
    • Materials and Methods
    • Acknowledgments
    • Footnotes
    • References
  • Figures & SI
  • Info & Metrics
  • PDF

You May Also be Interested in

Abstract depiction of a guitar and musical note
Science & Culture: At the nexus of music and medicine, some see disease treatments
Although the evidence is still limited, a growing body of research suggests music may have beneficial effects for diseases such as Parkinson’s.
Image credit: Shutterstock/agsandrew.
Scientist looking at an electronic tablet
Opinion: Standardizing gene product nomenclature—a call to action
Biomedical communities and journals need to standardize nomenclature of gene products to enhance accuracy in scientific and public communication.
Image credit: Shutterstock/greenbutterfly.
One red and one yellow modeled protein structures
Journal Club: Study reveals evolutionary origins of fold-switching protein
Shapeshifting designs could have wide-ranging pharmaceutical and biomedical applications in coming years.
Image credit: Acacia Dishman/Medical College of Wisconsin.
White and blue bird
Hazards of ozone pollution to birds
Amanda Rodewald, Ivan Rudik, and Catherine Kling talk about the hazards of ozone pollution to birds.
Listen
Past PodcastsSubscribe
Goats standing in a pin
Transplantation of sperm-producing stem cells
CRISPR-Cas9 gene editing can improve the effectiveness of spermatogonial stem cell transplantation in mice and livestock, a study finds.
Image credit: Jon M. Oatley.

Similar Articles

Site Logo
Powered by HighWire
  • Submit Manuscript
  • Twitter
  • Facebook
  • RSS Feeds
  • Email Alerts

Articles

  • Current Issue
  • Latest Articles
  • Archive

PNAS Portals

  • Anthropology
  • Chemistry
  • Classics
  • Front Matter
  • Physics
  • Sustainability Science
  • Teaching Resources

Information

  • Authors
  • Editorial Board
  • Reviewers
  • Librarians
  • Press
  • Site Map
  • PNAS Updates

Feedback    Privacy/Legal

Copyright © 2021 National Academy of Sciences. Online ISSN 1091-6490