JVI Accepts, published online ahead of print on 19 May 2010 J. Virol. doi: /jvi

Similar documents
MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells

Soft Agar Assay. For each cell pool, 100,000 cells were resuspended in 0.35% (w/v)

Determination of the temporal pattern and importance of BALF1 expression in Epstein-Barr viral infection

Chapter 4 Cellular Oncogenes ~ 4.6 -

Action and Mechanism of Epstein Barr virus Latent Membrane Protein1. induced Immortalization of Mouse Embryonic Fibroblasts *

MOLECULAR BASIS OF ONCOGENESIS

mirna Dr. S Hosseini-Asl

Epstein-Barr Virus BART MicroRNAs Are Produced from a Large Intron prior to Splicing

mir-509-5p and mir-1243 increase the sensitivity to gemcitabine by inhibiting

Epstein-Barr virus driven promoter hypermethylated genes in gastric cancer

Nanoparticles and persistent virus infection a dangerous liaison for the development of chronic lung disease(s)? Tobias Stöger

Epstein-Barr Virus BZLF1 Protein Binds to Mitotic Chromosomes

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Supplementary Materials for

EBV infection B cells and lymphomagenesis. Sridhar Chaganti

Supplementary Information. Supplementary Figure 1

Supplementary Figure 1. HOPX is hypermethylated in NPC. (a) Methylation levels of HOPX in Normal (n = 24) and NPC (n = 24) tissues from the

Herpesviruses. Virion. Genome. Genes and proteins. Viruses and hosts. Diseases. Distinctive characteristics

Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION

An Epstein-Barr virus-encoded microrna targets PUMA to promote host cell survival

Downregulation of MicroRNA-200 in EBV-Associated Gastric Carcinoma

Yamaguchi University School of Medicine, Kogushi, Ube, Yamaguchi. growth in low serum, anchorage-independent growth in soft

Supplementary Figure 1 IL-27 IL

Evaluation of the Lytic Origins of Replication of Kaposi s Sarcoma-Associated Virus/Human Herpesvirus 8 in the Context of the Viral Genome

Supplemental Materials and Methods Plasmids and viruses Quantitative Reverse Transcription PCR Generation of molecular standard for quantitative PCR

SUPPLEMENTARY INFORMATION. Supplementary Figures S1-S9. Supplementary Methods

Activation of cellular proto-oncogenes to oncogenes. How was active Ras identified?

Virus-host interactions

Transcription and RNA processing

Supplementary information

Publisher Rights Statement: Checked for eligibility: 11/08/2014. Copyright 2014, American Society for Microbiology. All Rights Reserved.

Received 2 November 2007/Accepted 19 February 2008

What causes cancer? Physical factors (radiation, ionization) Chemical factors (carcinogens) Biological factors (virus, bacteria, parasite)

EBV Tegument Protein BNRF1 Disrupts DAXX-ATRX to Activate Viral Early Gene Transcription

VIRUSES AND CANCER Michael Lea

Supplementary Figure 1

CELL CYCLE MOLECULAR BASIS OF ONCOGENESIS

on November 3, 2018 by guest

Oncolytic Viruses as a Potential Approach to Eliminate the HIV Reservoir

Recombinant Protein Expression Retroviral system

~Lentivirus production~

Functional characterisation of hepatitis B viral X protein/microrna-21 interaction in HBVassociated hepatocellular carcinoma

Supplementary Information

Where a licence is displayed above, please note the terms and conditions of the licence govern your use of this document.

BIT 120. Copy of Cancer/HIV Lecture

Viruses Tomasz Kordula, Ph.D.

Supplemental Figure 1

Viruses. Rotavirus (causes stomach flu) HIV virus

MicroRNA dysregulation in cancer. Systems Plant Microbiology Hyun-Hee Lee

Cancer genetics

Lentiviral Delivery of Combinatorial mirna Expression Constructs Provides Efficient Target Gene Repression.

Transcription and RNA processing

Supplemental Table S1

John Bell Centre for Innovative Cancer Therapeutics

of Nebraska - Lincoln

Egr-1 regulates RTA transcription through a cooperative involvement of transcriptional regulators

Part-4. Cell cycle regulatory protein 5 (Cdk5) A novel target of ERK in Carb induced cell death

SUPPLEMENTARY INFORMATION

Epstein-Barr Virus (EBV) SM Protein Induces and Recruits Cellular Sp110b To Stabilize mrnas and Enhance EBV Lytic Gene Expression

HEK293FT cells were transiently transfected with reporters, N3-ICD construct and

Supplementary information. MARCH8 inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins

PERSISTENCE OF EBV IN THE CANCER STEM CELL FRACTION OF MULTIPLE MYELOMA

Doctoral Degree Program in Marine Biotechnology, College of Marine Sciences, Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei,

HIF-inducible mir-191 promotes migration in breast cancer through complex regulation of TGFβ-signaling in hypoxic microenvironment.

Supplementary Fig. 1. Delivery of mirnas via Red Fluorescent Protein.

Epstein-Barr Virus: Cell Trafficking Is Crucial for Persistence

s u p p l e m e n ta ry i n f o r m at i o n

Supplementary Information Titles Journal: Nature Medicine

Stage-Specific Inhibition of MHC Class I Presentation by the Epstein-Barr Virus BNLF2a Protein during Virus Lytic Cycle

Epstein-Barr Virus: Stimulation By 5 '-Iododeoxy uridine or 5 '-Brom odeoxy uridine in Human Lymphoblastoid Cells F ro m a Rhabdom yosarcom a*

The Biology and Genetics of Cells and Organisms The Biology of Cancer

EBV Infection and Immunity. Andrew Hislop Institute for Cancer Studies University of Birmingham

Restricted VZV transcription in human trigeminal ganglia

Table S1. Primer sequences used for qrt-pcr. CACCATTGGCAATGAGCGGTTC AGGTCTTTGCGGATGTCCACGT ACTB AAGTCCATGTGCTGGCAGCACT ATCACCACTCCGAAGTCCGTCT LCOR

VIROLOGY. Engineering Viral Genomes: Retrovirus Vectors

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

Inhibition of mtorc1 inhibits lytic replication of Epstein-Barr virus in a cell-type specific manner

An epithelial-to-mesenchymal transition-inducing potential of. granulocyte macrophage colony-stimulating factor in colon. cancer

EBV essential antigen EBNA3C attenuates H2AX expression

Received 16 January 2009/Accepted 5 May 2009

Chronic Viral Infections vs. Our Immune System: Revisiting our view of viruses as pathogens

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath

Lytic Cycle Switches of Oncogenic Human Gammaherpesviruses 1

Viruses. Poxviridae. DNA viruses: 6 families. Herpesviridae Adenoviridae. Hepadnaviridae Papovaviridae Parvoviridae

Serum mir-200c expression level as a prognostic biomarker for gastric cancer

Cancer. Questions about cancer. What is cancer? What causes unregulated cell growth? What regulates cell growth? What causes DNA damage?

Oncogenes. Dr. S Hosseini-Asl

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved.

Fayth K. Yoshimura, Ph.D. September 7, of 7 RETROVIRUSES. 2. HTLV-II causes hairy T-cell leukemia

VIII Curso Internacional del PIRRECV. Some molecular mechanisms of cancer

Supplementary Figure 1. SC35M polymerase activity in the presence of Bat or SC35M NP encoded from the phw2000 rescue plasmid.

Arsenic trioxide inhibits EBV reactivation and promotes cell death in EBV-positive lymphoma cells

Activation of Gene Expression by Human Herpes Virus 6

Type of file: PDF Size of file: 0 KB Title of file for HTML: Supplementary Information Description: Supplementary Figures

Ubiquitination and deubiquitination of NP protein regulates influenza A virus RNA replication

Use of the red fluorescent protein as a marker of Kaposi s sarcoma-associated herpesvirus lytic gene expression

Bmi-1 regulates stem cell-like properties of gastric cancer cells via modulating mirnas

Leflunomide/teriflunomide inhibit Epstein-Barr virus (EBV)-induced lymphoproliferative disease and lytic viral replication

Inventory of Supplemental Data: Supplemental Figures 1-7. Supplemental Tables 1-8. Supplemental References. Supplemental Video:

Stability determinants of Murine Cytomegalovirus long non-coding RNA7.2

Transcription:

JVI Accepts, published online ahead of print on 19 May 2010 J. Virol. doi:10.1128/jvi.00379-10 Copyright 2010, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved. Title: Differential expression of the mir-200 family micrornas in epithelial and B-cells and regulation of Epstein-Barr virus reactivation by the mir-200 family member, mir- 429. Running title: Regulation of EBV reactivation by mir-429 Authors: Zhen Lin, Xia Wang, Claire Fewell, Jennifer Cameron, Qinyan Yin, and Erik K Flemington* Address: Department of Pathology, SL-79 Tulane Health Sciences Center 1430 Tulane Avenue New Orleans, Louisiana 70112 USA *Corresponding Author: Erik K Flemington Tel: 504-988-1167 Email: eflemin@tulane.edu Word counts Abstract: 100 Text: 1814 (title page through acknowledgements) 1

Abstract: The mir-200 microrna family is important for maintaining epithelial phenotype, partially through suppressing ZEB1 and ZEB2. Since ZEB1 inhibits EBV reactivation, we hypothesized that expression of mir-200 family members in epithelial cells may partly account for higher EBV reactivation levels in this tissue (relative to non-plasma B-cells). Here we show that, whereas mir-200 family members are expressed in epithelial cells, their expression is low in latently infected B-cells. Further, the mir-200 family member, mir-429, shows elevated expression in plasma-cell lines and is induced by B-cellreceptor activation in Akata cells. Lastly, expression of mir-429 can break latency. Downloaded from http://jvi.asm.org/ on October 15, 2018 by guest 2

Epstein-Barr virus (EBV) is an important human pathogen that is involved in a variety of malignant and non-malignant diseases (16). EBV utilizes two distinct stages in its lifecycle: latency and a viral replication stage (lytic reactivation). Expression of the viral lytic gene products, Zta or Rta, initializes the entire cascade of lytic gene expression, leading to production of infectious virus. The switch mechanism controlling the transition from latency to the lytic replication phase is tightly regulated to maintain a homeostatic viral life-cycle. A number of cellular and viral factors have been identified that regulate the Zta and Rta promoters. Kraus et al (11) have shown that the EMT regulator, ZEB1, can bind to an inhibitory element in the Zta promoter named ZV. The binding of ZEB1 to ZV inhibits the reactivation process by blocking the expression of Zta (8, 11, 20). Further, a recombinant EB-virus with a disrupted ZV element displays a higher reactivation rate and higher yields of infectious virions than wild-type virus (20). MicroRNAs are involved in regulating a wide spectrum of cellular processes as well as the life-cycle of viruses. Herpesviruses encode their own sets of micrornas that modulate host signaling pathways in ways that promote their virus infection cycle (3-5). Viruses also utilize host cell mirnas to modulate the cellular environment in ways that similarly support their infection cycle (18). The mir-200 family of micrornas function in various biological and pathological processes and there is accumulating evidence that they can function as potent tumor suppressors (15). This family has 5 members that are located in two distinct genomic loci/clusters (Figure 1A). One cluster is located on chromosome 1 and encodes mir- 200a, mir-200b, and mir-429. The second cluster is located on chromosome 12 and encodes mir-200c and mir-141. Members of each cluster are largely co-regulated as 3

a result of being processed from the same primary transcript. The mir-200 family can also be divided into two presumed functional groups based on their seed sequences (mir-429, mir-200b, and mir-200c have identical seed sequences that differ by 1 base with the mir-200a and mir-141 seeds). Although a one base difference in seed sequence is expected to confer largely distinct targeting specificity, Park et al (14) have shown that these subgroups have highly overlapping targetomes, suggesting that they have evolutionarily conserved functions. Expression of the mir-200 family mirnas in EBV-trophic cell lines. The level of mir-200 family expression is commonly altered in both physiological (e.g. EMT and embryogenesis) and pathological (e.g. tumorigenesis) processes. As a direct target of the mir-200 family, ZEB1 shows an inverse expression pattern with the mir-200 family (14). Considering the fact that ZEB1 serves as a master regulator for EBV reactivation, we tested the expression of mir-200 family mirnas in tissue types that are naturally permissive for EBV infection. Total RNA was extracted from EBV-negative and EBVpositive (type I or type III latency) B-cell lines (Burkitt s lymphomas and lymphoblastoid cell lines) and epithelial cells using a modified TRIzol method (2) and a mirneasy kit (Qiagen). Quantitative RT-PCR analysis of the mir-200b~200a~429 primary transcript revealed little expression in each B-cell line tested (Figure 1A and 1B). In contrast, expression of this cluster was readily detected in all EBV-positive and EBV-negative epithelial cell lines (Figure 1B). We then examined expression of the mature/processed form of a representative mir-200 family member from this locus, mir-429, using a TaqMan mirna assay (Applied Biosystems). Again, while expression was observed in all epithelial cell lines, little or no expression was detected in any of these B-cell lines 4

(Figure 1C). Lastly, a member of the second mir-200 family locus, mir-200c, was similarly found to be substantially lower in each B-cell line compared to epithelial cells (Figure 1D). Induction of EBV lytic reactivation in mir-429 transduced EBV-293 cells. To investigate the influence of mir-200 family members on reactivation, we introduced a control or a mir-429 expressing pmscv-puro-gfp-mir (2) retrovirus (in triplicate) into EBV-infected 293 fibroblastic cells (EBV-293). A higher level of mature mir-429 was detected in all mir-429 transduced cell cultures compared to control transduced cultures (Figure 2A). Consistent with previous studies showing that mir-200 micrornas can function as master regulators of EMT, mir-429 transduced EBV-293 cells lost their spindle-shaped cell morphology and acquired clustered growth characteristics with cobble-stone shaped appearance (Figure 2B). Western blot analysis of ZEB1 expression (anti-zeb1 polyclonal antibody (Santa Cruz, cat# sc- 25388)) showed the expected reduction in mir-429 transduced cells (Figure 2C) (antiactin polycolonal antibody (Santa Cruz, cat# sc-1615)). This indicated to us that the level of mir-429 expressed in cells transduced with the mir-429 retrovirus was sufficient to elicit an expected change in phenotype and the expected suppression of a mir-429 target. We then assessed the number of Zta positive cells in mir-429 and control transduced cultures by immunofluorescence following staining with an anti-zta antibody (Argene, cat# 11-007) and a goat-anti-mouse Alexa Fluor 594 (Invitrogen cat# A11032) secondary antibody. A greater number of Zta positive cells were observed in all three mir-429 transduced cell cultures compared to control transduced cultures (Figure 2D) 5

indicating that this mir-200 family member can shift the latent/lytic balance towards the lytic cycle in EBV-293 cells. Induction of EBV lytic reactivation in mir-429 transduced B-cell lines. Next, we examined whether mir-429 could disrupt latency in B-cells. Mutu I cells transduced with the mir-429 retrovirus showed higher mature mir-429 expression in all three infections compared to cells transduced with the control retrovirus, with a concordant reduction in ZEB1 levels (Figure 3A). Western blot analysis of the immediate early and early genes, Zta (Argene, cat# 11-007), Rta (Argene, cat#11-008), and BMRF1 (Capricorn, cat# EBV-018-48180) showed increased expression of all three of these viral genes in mir-429 transduced cells (Figure 3A) (fourteen days after selection). Similarly, introduction of a mir-429 retrovirus into another EBV positive B-cell line, Akata, induced Zta, Rta, and BMRF1 expression (Figure 3B). Together, these results demonstrate that mir-429 can also induce EBV reactivation in B-cells. mir-429 is induced by B-cell receptor activation and shows increased expression in plasma cell lines. In vivo, EBV reactivation in B-cells is thought to occur through B- cell differentiation and high lytic activity is observed in tonsillar plasma cells (6). Synchronous EBV reactivation can be achieved in tissue culture through activation of the B-cell receptor (BCR) (17). BCR activation in Akata cells by treatment with anti-ig (Sigma, cat # I5260) resulted in a 113 fold increase in mature mir-429 levels (Figure 3C). Analysis of mir-429 expression in two plasma cell lines showed elevated levels relative to the expression observed in Mutu I or unstimulated Akata cells (Figure 3C). Together, these data suggests that in addition to playing a possible role in supporting 6

lytic replication in epithelial cells, the mir-200 family may support BCR mediated reactivation in B-cells and reactivation in plasma cells. mir-429 induces the Zta promoter. To investigate whether mir-429 can induce reactivation in part through the Zta promoter (Zp), we carried out a reporter experiment in 293 cells infected with a Zta knockout EB virus (EBV(Zk/o)/293, (7)). 10 cm plates of EBV(Zk/o)/293 cells were co-transfected with 5 ug of a control or a mir-429 expression vector plus 2 ug of a luciferase reporter plasmid containing Zp sequences (-221 to +13 (10), which contains the previously identified ZEB1 binding site (11)). In this setting, mir-429 induced Zp activity 1.8 fold (Figure 4A). In the presence of 0.5 ug of a cotransfected Zta expression vector (to facilitate an activated state of the Zta promoter (9, 12)), mir-429 induced Zp activity 2.4 fold (Figure 4A). We then mutated the ZV-ZEB1 binding element (using a site-directed mutagenesis kit, Stratagene, cat# 200519) in the Zp reporter plasmid and tested whether this abrogates responsiveness to mir-429. A statistically significant reduction in responsiveness to mir-429 was observed but it nevertheless remained responsive to mir-429 (Figure 4B and 4C). The mutation that we introduced in our construct was shown previously to largely abrogate ZEB1 binding to this element (8, 11) so it is unlikely that the remaining response is due to residual binding of ZEB1 to this element. These results indicate that mir-429 induces the Zta promoter through the ZV-ZEB1 binding site but that there are likely additional elements/factors that help facilitate induction of the Zta promoter. We propose that the mir-200 family, which is a critical microrna determinant of epithelial cell phenotype, may be a contributing effector of EBV s propensity to exhibit a bias toward the lytic phase in epithelial cells. On the other end of the spectrum, we 7

observe little if any mir-200 family expression in any non-plasma B-cell line tested, consistent with the generally more latent bias observed in this setting. The findings that mir-429 expression is elevated in plasma cells and following BCR engagement suggests that mir-200 family members may also be involved in supporting reactivation in B-cell settings where viral reactivation occurs. It is also notable that HDAC inhibitors, which are known to induce EBV reactivation, have been shown to induce mir-200 micrornas (21). It is conceivable that HDAC inhibitors may induce reactivation in part through their influence on this pathway. Lastly, while the ZV-ZEB1 binding site in Zp may mediate the influence of mir-429 on Zp, induction of reactivation is likely to be more complex. There may be other uncharacterized ZEB1 binding sites within Zp and there may be other Zp binding factors that are influenced by mir-429. Lastly, ZEB1 has been shown to bind to the Rta promoter (11, 20) raising the possibility that the respective binding site(s) may similarly be responsive to mir-200 family members and thereby contribute to mir-429 mediated reactivation. Acknowledgements: This work was supported by the NIH grants, CA124311, CA130752, and CA138268 EKF. EBV-293 and EBV(Zk/o)/293 cells were a generous gift from Henri-Jacques Delecluse and Luwen Zhang. 8

Figure Legends: Figure 1. Expression of mir-200 family mirnas in EBV-trophic cell lines. (A) mir-200 family genomic loci. mir-200b, mir-200a, and mir-429 are transcribed as a single polycistronic transcript from chromosome 1. mir-200c and mir-141 are encoded by a single transcript from chromosome 12. Chromosomal coordinates are based on the 2009 version of the public human genome assembly (GRCh37). (B) Quantitative RT-PCR of primary mir-200b~200a~429 transcript. Primer set for Pri-miR-200b~200a~429: 5 -AGTGGGGCTCACTCTCCAC-3 and 5 - AGGAGGAGGAGGAGGAGAAA-3 (1). Primer set for G3PDH: 5 - GCCAAGGTCATCCATGACAACTTTGG-3, and 5 - GCCTGCTTCACCACCTTCTTGATGTC-3. The expression of Primary mir- 200b~200a~429 was determined by the comparative CT method (2 CT ). Mutu I EBV positive type I latency Burkitt s lymphoma cell line; JY, Jijoye, and X50-7 EBV positive type III latency B-cell lines; OKF6/TERT2 EBV negative telomerase immortalized human keratinocyte cell line; CNE1 and CNE1-EBV EBV negative and positive nasopharyngeal carcinoma cell lines; AGS EBV negative human gastric adenocarcinoma cell line; MCF7 EBV negative human breast cancer cell line. (C) Mature mir-429 expression was quantified using a TaqMan microrna assay (Applied Biosystems). RNU6B was analyzed as a reference. The expression of mir-429 was determined by the comparative C T method (2 CT ). DG75 EBV negative Burkitt s lymphoma cell line; Rael and Akata EBV positive type I 9

latency Burkitt s lymphoma cell lines; AGS-EBV AGS cells infected with a recombinant EBV virus. Other cells are as in panel B legend. (D) Mature mir-200c expression was assessed using a mirvana (qrt-pcr) microrna detection kit (Ambion, cat# AM1558) with the mirvana qrt-pcr mir- 200c primer set (Ambion, cat# AM30096) and the mirvana qrt-pcr 5S primer set (Ambion, cat# AM30302) according to the manufacturer's protocol. The expression of mir-200c was determined by the comparative C T method (2 CT ). Cell lines are defined in panel B and C legends. Figure 2. Induction of EBV lytic reactivation in mir-429 transduced EBV-293 cells. (A) A 441-bp region spanning the mir-429 hairpin sequence (chr1:1,104,265-1,104,705 of GRCH37 assembly) was isolated from JY genomic DNA by PCR and cloned into the plasmid, pmscv-puro-gfp-mir (2). Retroviral infection was conducted as described previously (19) and cells were selected with puromycin for 14 days prior to analysis. Mature mir-429 expression was analyzed by quantitative RT-PCR and values were determined by the comparative C T method (2 CT ) using RNU6B as a control. (B) Two separate mir-429 transduced EBV-293 cell cultures show morphological changes relative to two separate control transduced EBV-293 cell cultures. (C) Western blot analysis of ZEB1 expression in retrovirally transduced EBV-293 cells. 10

(D) Immunofluorescence assay of Zta in retrovirally transduced EBV-293 cells. The assay was performed as previously described (13). The number of Zta positive cells on coverslips containing 12,000 total cells were counted for each culture. The average and error from triplicate coverslips are shown for each cell culture. Figure 3. Induction of EBV reactivation in mir-429 transduced B-cell lines. (A) Generation of retrovirally transduced Mutu I cell lines and analysis of mature mir-429 expression were performed as described in figure legend 2A. ZEB1 and actin were detected using polyclonal antibodies as in figure 2C. (B) Retrovirally transduced Akata cell lines were generated in the same manner as the Mutu I cell lines and analysis of mature mir-429 expression was performed as described in figure legend 2A. ZEB1 and actin were detected using polyclonal antibodies as in figure 2C. (C) Mature mir-429 expression was quantified using a TaqMan microrna assay (Applied Biosystems). RNU6B was analyzed as a reference. The expression of mir-429 was determined by the comparative C T method (2 CT ). Akata cells were treated with anti-human Ig (Sigma I5260) and cells were harvested at 0 hour, 8 hours, 12 hours and 24 hours post-treatment. Human plasma cell lines U266 and CCL-155 were obtained from ATCC. Figure 4. mir-429 induces the Zta promoter. were co-transfected with either a mir-429 expression vector (pmscv-puro-gfp- (A) 293 cells stably infected with a Zta knockout EB virus (EBV(Zk/o)/293 (7)) were transfected using a modified calcium phosphate precipitation method (13). Cells 11

mir-429) or a control vector (pmscv-puro-gfp-mircntl) plus either a Zta expression vector (BS(SVp/e)-Zta) or its control vector (BS(SVp/e)). Cells were harvested 72 hours after transfection, reporter activity was analyzed by a luciferase assay, and protein levels were assessed by Western blot analysis. Fold changes are relative to cells transfected with Zta control vector plus mir- 429 control vector. (B) EBV(Zk/o)/293 cells were transfected with the indicated plasmids. Zp-Luc is a luciferase reporter plasmid containing Zp sequences (-221 to +13). ZVmut-Luc is a luciferase reporter plasmid bearing a defective ZV-ZEB1 binding site that was generated by changing the A at position -12 (relative to the Zp transcriptional initiation site) to a C. BG-Luc, which contains a minimal beta-globin promoter, was used as a control reporter construct. RLU: relative light units. (C) Fold changes are relative to Zta induced reporter activity in the absence of mir- 429. Statistical significance of the difference of fold change between Zp-Luc group and ZVmut-Luc group was analyzed by the student s t-test method. 12

References: 1. Bracken, C. P., P. A. Gregory, N. Kolesnikoff, A. G. Bert, J. Wang, M. F. Shannon, and G. J. Goodall. 2008. A double-negative feedback loop between ZEB1-SIP1 and the microrna-200 family regulates epithelial-mesenchymal transition. Cancer Res 68:7846-54. 2. Cameron, J. E., Q. Yin, C. Fewell, M. Lacey, J. McBride, X. Wang, Z. Lin, B. C. Schaefer, and E. K. Flemington. 2008. Epstein-Barr virus latent membrane protein 1 induces cellular MicroRNA mir-146a, a modulator of lymphocyte signaling pathways. J Virol 82:1946-58. 3. Cullen, B. R. 2009. Viral and cellular messenger RNA targets of viral micrornas. Nature 457:421-5. 4. Cullen, B. R. 2009. Viral RNAs: lessons from the enemy. Cell 136:592-7. 5. Cullen, B. R. 2006. Viruses and micrornas. Nat Genet 38 Suppl:S25-30. 6. dlaichalk, L. L., and D. A. Thorley-Lawson. 2005. Terminal differentiation into plasma cells initiates the replicative cycle of Epstein-Barr virus in vivo. J Virol 79:1296-307. 7. Feederle, R., M. Kost, M. Baumann, A. Janz, E. Drouet, W. Hammerschmidt, and H. J. Delecluse. 2000. The Epstein-Barr virus lytic program is controlled by the co-operative functions of two transactivators. EMBO J 19:3080-9. 8. Feng, W. H., R. J. Kraus, S. J. Dickerson, H. J. Lim, R. J. Jones, X. Yu, J. E. Mertz, and S. C. Kenney. 2007. ZEB1 and c-jun levels contribute to the establishment of highly lytic Epstein-Barr virus infection in gastric AGS cells. J Virol 81:10113-22. 13

9. Flemington, E., and S. H. Speck. 1990. Autoregulation of Epstein-Barr virus putative lytic switch gene BZLF1. J Virol 64:1227-32. 10. Flemington, E., and S. H. Speck. 1990. Identification of phorbol ester response elements in the promoter of Epstein-Barr virus putative lytic switch gene BZLF1. J Virol 64:1217-26. 11. Kraus, R. J., J. G. Perrigoue, and J. E. Mertz. 2003. ZEB negatively regulates the lytic-switch BZLF1 gene promoter of Epstein-Barr virus. J Virol 77:199-207. 12. Lieberman, P. M., and A. J. Berk. 1990. In vitro transcriptional activation, dimerization, and DNA-binding specificity of the Epstein-Barr virus Zta protein. J Virol 64:2560-8. 13. Lin, Z., Q. Yin, and E. Flemington. 2004. Identification of a negative regulatory element in the Epstein-Barr virus Zta transactivation domain that is regulated by the cell cycle control factors c-myc and E2F1. J Virol 78:11962-71. 14. Park, S. M., A. B. Gaur, E. Lengyel, and M. E. Peter. 2008. The mir-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev 22:894-907. 15. Peter, M. E. 2009. Let-7 and mir-200 micrornas: guardians against pluripotency and cancer progression. Cell Cycle 8:843-52. 16. Rickinson, A. B., and E. Kieff. 2007. Epstein-Barr virus. In: D.M. Knipe and P.M. Howley, Editors, Fields Virology (5th ed.), Lippincott Williams & Wilkins, Philadelphia, PA.:2655-2700. 17. Takada, K., and Y. Ono. 1989. Synchronous and sequential activation of latently infected Epstein-Barr virus genomes. J Virol 63:445-9. 14

18. Umbach, J. L., and B. R. Cullen. 2009. The role of RNAi and micrornas in animal virus replication and antiviral immunity. Genes Dev 23:1151-64. 19. Yin, Q., J. McBride, C. Fewell, M. Lacey, X. Wang, Z. Lin, J. Cameron, and E. K. Flemington. 2008. MicroRNA-155 is an Epstein-Barr virus-induced gene that modulates Epstein-Barr virus-regulated gene expression pathways. J Virol 82:5295-306. 20. Yu, X., Z. Wang, and J. E. Mertz. 2007. ZEB1 regulates the latent-lytic switch in infection by Epstein-Barr virus. PLoS Pathog 3:e194. 21. Zhang, S., X. Cai, F. Huang, W. Zhong, and Z. Yu. 2008. Effect of trichostatin a on viability and microrna expression in human pancreatic cancer cell line BxPC- 3. Exp Oncol 30:265-8. Downloaded from http://jvi.asm.org/ on October 15, 2018 by guest 15