Free access
Research Article
15 November 2010

Virally Induced Cellular MicroRNA miR-155 Plays a Key Role in B-Cell Immortalization by Epstein-Barr Virus

ABSTRACT

Infection of resting primary human B cells by Epstein-Barr virus (EBV) results in their transformation into indefinitely proliferating lymphoblastoid cell lines (LCLs). LCL formation serves as a model for lymphomagenesis, and LCLs are phenotypically similar to EBV-positive diffuse large B-cell lymphomas (DLBCLs), which represent a common AIDS-associated malignancy. B-cell infection by EBV induces the expression of several cellular microRNAs (miRNAs), most notably miR-155, which is overexpressed in many tumors and can induce B-cell lymphomas when overexpressed in animals. Here, we demonstrate that miR-155 is the most highly expressed miRNA in LCLs and that the selective inhibition of miR-155 function specifically inhibits the growth of both LCLs and the DLBCL cell line IBL-1. Cells lacking miR-155 are inefficient in progressing through S phase and spontaneously undergo apoptosis. In contrast, three other B-cell lymphoma lines, including two EBV-positive Burkitt's lymphoma cell lines, grew normally in the absence of miR-155 function. These data identify the induction of cellular miR-155 expression by EBV as critical for the growth of both laboratory-generated LCLs and naturally occurring DLBCLs and suggest that targeted inhibition of miR-155 function could represent a novel approach to the treatment of DLBCL in vivo.
Epstein-Barr virus (EBV) is a human gammaherpesvirus that is able to establish a long-term, latent infection in human B cells for the life of the host (40). EBV infection is associated with a range of human cancers, including Burkitt's lymphoma (BL) and Hodgkin's disease, as well as several AIDS-associated cancers, of which the most prevalent is diffuse large B-cell lymphoma (DLBCL) (7). It has been proposed that EBV initially infects naïve B cells and then induces these to undergo a period of rapid proliferation leading eventually to differentiation into a pool of latently EBV-infected cells that resemble memory B cells (45). These two successive phases are referred to as EBV latency III and EBV latency I. During latency III, EBV expresses a set of nine proteins—the EBV nuclear antigen (EBNA) proteins EBNA-1, -2, -3A, -3B, -3C, and -LP and the latent membrane proteins (LMPs) LMP-1, -2A, and -2B—that are thought to mimic the ordered activation process that a naïve B cell would normally undergo after antigenic stimulation (22, 45). Eventually, these cells cycle back into a differentiation state that resembles that of a memory B cell, at which point EBV protein expression becomes restricted to EBNA-1.
Previously published models for EBV latency proposed that the reprogramming of B cells after EBV infection largely reflected the action of viral proteins (22). These were thought to modify the pattern of cellular gene expression either directly, by acting as transcription factors, or indirectly, by mimicking the signals normally generated at the cell surface during antigenic stimulation. However, recent data have also revealed that EBV encodes no fewer than 25 distinct microRNAs (miRNAs) that have the potential to also play a critical role in the establishment and/or maintenance of EBV latency (5, 19, 36, 56). Moreover, it is now clear that EBV infection greatly perturbs not only the pattern of cellular mRNA expression in infected B cells but also the pattern of cellular miRNA expression. In particular, EBV infection of resting primary B cells has been shown to strongly activate the expression of miR-155 and a small number of other cellular miRNAs (6, 15, 21, 32).
miRNAs are a class of ∼22-nucleotide (nt)-long RNAs that function as negative posttranscriptional regulators of gene expression by binding to complementary sites located on target mRNAs (2). Particularly critical for efficient downregulation of mRNA expression is the miRNA seed region, nucleotides 2 to 8 from the 5′ end of the miRNA, which generally must base pair to a fully complementary mRNA sequence. The human genome encodes >700 distinct miRNAs, and miRNAs are also encoded by a range of different herpesviruses, including EBV and the related gammaherpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV) (47). While most viral miRNAs lack sequence homology to known cellular miRNAs, there is an exception in the case of KSHV miR-K11, which has the same seed sequence as cellular miR-155 (18, 42). Moreover, a second transforming herpesvirus, the avian alphaherpesvirus Marek's disease virus (MDV), also encodes an miRNA, miR-M4, with full seed homology to miR-155 (31, 55). Importantly, analysis of a number of candidate miR-155 mRNA targets has revealed that cellular miR-155, KSHV miR-K11, and MDV miR-M4 are all able to downregulate these mRNAs equivalently, thus validating miR-K11 and miR-M4 as true functional orthologs of miR-155 (18, 42, 55). As miR-155 overexpression has been shown to cause tumors in mice as well as in chickens (8, 9) and high levels of miR-155 expression are observed in many forms of B-cell lymphoma (13, 24, 51), the presence of miR-155 orthologs in these two herpesviruses may imply a role in viral tumorigenesis.
Although EBV does not itself encode an miRNA with homology to miR-155, EBV infection does, as noted above, strongly activate cellular miR-155 expression (6, 15, 21, 32). It therefore seemed possible that overexpression of miR-155 of either cellular origin (in the case of EBV) or viral origin (in the case of KSHV and MDV) might play a key role in the life cycle of all these viruses, perhaps by promoting the activation and growth of infected lymphocytes. Previous studies have shown that miR-155 can target mRNAs that function in key cell signal transduction pathways (27, 39, 52) and cell cycle regulatory pathways (4). Specifically, in the context of EBV, miR-155 was shown to be important for the efficient episomal maintenance of the EBV genome (27), and in the context of reticuloendotheliosis virus, it was shown to be important for cell survival (4). Here, we demonstrate that the ablation of miR-155 function in the context of EBV-infected primary B cells indeed blocks their normally rapid proliferation and increases their tendency to undergo apoptosis.

MATERIALS AND METHODS

DNA constructs.

The indicator vector pNL-SIN-CMV-FLuc, which was used as an internal control for detection of miRNA function, has been previously described, as has pNL-SIN-CMV-RLuc (16). A derivative of pNL-SIN-CMV-RLuc bearing two fully complementary artificial target sites for cellular miR-155 (1552T) has also been described previously (18). For indicator assays for lymphoblastoid cell lines (LCLs), the segment of DNA encoding firefly luciferase (FLuc) or Renilla luciferase (RLuc)-1552T was removed from the pNL-SIN-CMV constructs and inserted into pLCE—the same backbone used for the lentiviral sponge constructs—using restriction sites NheI and XbaI. The resultant plasmids were named pLC-FLuc and pLC-RLuc-1552T. A lentiviral sponge specific for miR-155 was constructed as described previously (12, 17), and the control green fluorescent protein (GFP) and sCXCR4 vectors were published previously (17). For this study, a nine-copy sponge sequence specific for miR-155 (s155) was cloned into lentiviral vector pLCE as described previously (17) by using oligonucleotides 5′-CTAGGACCCCTATCACACCTAGCATTAAGTTTGACCCCTATCACACCTAGCATTAAGTTTGACCCCTATCACACCTAGCATTAATCTAGATTTGAATTC-3′ and 5′-AATTGAATTCAAATCTAGATTAATGCTAGGTGTGATAGGGGTCAAACTTAATGCTAGGTGTGATAGGGGTCAAACTTAATGCTAGGTGTGATAGGGGTC-3′.
The pMSCV/GFP and pMSV/s155 retroviral vectors were generated from pMSCV-puro (catalog no. 634401; Clontech) by excision of the GFP and GFP-s155 expression cassettes from the relevant pLCE-based vector by cleavage with NheI and EcoRI, followed by ligation of the resultant fragments into pMSCV-puro that had been cleaved with HpaI and EcoRI. Similarly, the pTRIPZ/GFP and pTRIPZ/s155 lentiviral vectors were generated from the tetracycline-regulatable vector pTRIPZ-puro (catalog no. RHS4696; Open Biosystems) by excision of the GFP and GFP-s155 expression cassettes from the pLCE-based vectors by cleavage with AgeI and EcoRI, followed by insertion of the resultant fragments into pTRIPZ-puro that had also been cleaved with AgeI and EcoRI.

cDNA library preparation and data analysis.

cDNA libraries for Solexa/Illumina sequencing were prepared as previously described (46). Briefly, small RNAs were isolated by preparative gel electrophoresis and sequentially ligated to 3′ and then 5′ linkers. Primers complementary to the linker sequences were used for reverse transcription (RT) and PCR in order to generate cDNA libraries for deep sequencing. Raw sequencing data were filtered to remove reads lacking identifiable 3′ linker sequences and/or reads falling outside the predicted miRNA size range. A list of final usable reads was then collapsed down into a list of unique sequences which was aligned to the cellular and viral pre-miRNA database from miRBase (release 14.0) by using NCBI BLAST and further parsed using the blastoutparse and filter_alignment scripts of the miRDeep software package (14).

Cell culture, generation of LCLs, and generation of viral transductants.

The EBV-positive BL cell lines Mutu I, Mutu III, Namalwa, and Raji, the EBV-positive DLBCL cell line IBL-1, and the EBV-negative BJAB cell line were all maintained in RPMI 1640 supplemented with 10% fetal bovine serum (FBS). EBV-infected human peripheral blood mononuclear cells (PBMCs) were cultured in RPMI 1640 with 15% FBS, and 293T cells were cultured in Dulbecco's modified Eagle's medium (DMEM) with 10% FBS. All cells were maintained in the presence of antibiotics.
The LCLs SDLCL, EF3D, and LCLd1 were generated by infection of PBMCs with EBV strain B95-8. PBMCs were isolated from buffy coats of normal donors (Carolina Red Cross) by using a Histopaque-1077 column (Sigma). A total of 107 PBMCs were infected with 500 μl of filtered B95-8 virus stock in the presence of 0.5 μg/ml cyclosporine in R15 medium (RPMI 1640 with 15% FBS and 50 μg/ml gentamicin) for 1 h at 37°C. For outgrowth, infected cells were brought to 14.4 ml in R15 medium plus cyclosporine (0.5 μg/ml) and plated in either a 24-well plate for time course analyses (one well was harvested for each time point) and the formation of SDLCL and LCLd1 or a 96-well plate for clonal expansion to generate EF3D.
Lentiviral particles were produced by cotransfection of the pLCE vectors with pMDLgpRRE, pRSV-Rev, and pVSV-G (11) into 293T cells by using Fugene6. pNL-SIN constructs were also packaged in 293T cells but by cotransfection with pcRev, pcTat, and pHIT/G (30). pMSCV was packaged using mouse stem cell virus (MSCV) Gag/Pol and pHIT/G; pTRIPZ was packaged using pCMVΔR8.75 and pMD2G. Supernatants were collected, filtered, and concentrated with Amicon Ultra centrifugal filters (Millipore) 48 h after transfection. The lentivirus- or retrovirus-containing supernatants were then used to transduce the cell lines for which growth curves are shown in Fig. 2. For LCLs, transduction efficiency was approximately 30 to 50%; for all other cell lines except for IBL-1, it was 80 to 90%. MSCV transduction of IBL-1 was only ∼15% efficient; therefore, IBL-1 cells were grown in the presence of puromycin for 1 week to select for transduced cells. Without sorting, all transduced cell populations were a mixture of GFP-positive and GFP-negative cells.

Indicator assays.

Indicator assays for miRNA activity were performed using previously described lentiviral RLuc- and FLuc-based indicator vectors (16). Briefly, pNL-SIN-CMV-FLuc and either pNL-SIN-CMV-RLuc or pNL-SIN-CMV-RLuc-1552T were packaged into viral particles and then cotransduced (FLuc plus RLuc) into control 293T cells or target B cells. Two days posttransduction, cells were collected and lysed and both FLuc and RLuc levels were quantified using a dual luciferase reporter assay (Promega). FLuc/RLuc ratios were calculated and normalized to ratios obtained from 293T cells not expressing miR-155. For the inducible system, miRNA indicator vectors were transduced into LCLd1/GFP and LCLd1/s155 cells after these had been expanded in the presence of puromycin and the absence of doxycycline. After transduction, the cells were washed and resuspended in media containing doxycycline. GFP-positive cells were sorted 2 days later and immediately lysed for quantitation of luciferase (both RLuc and FLuc) as described above.

Fluorescence-activated cell sorter (FACS) experiments.

LCLs (EF3D and SDLCL) growing as mixed populations of GFP-positive cells (lacking miR-155 function) and GFP-negative cells (retaining miR-155 function) were split to 2 × 105 cells/ml after lentiviral transduction as described above. Twenty-four hours later, while at equal concentrations and while still subconfluent, cells were labeled for 45 min with 25 μM 5-bromodeoxyuridine (BrdU; Calbiochem) and then collected. Cells were fixed with 4% paraformaldehyde on ice for 1 h, pelleted, and then washed twice with phosphate-buffered saline (PBS). Next, cells were permeabilized at room temperature in 0.5% Triton X-100 in PBS (PBS-T). After the cells were washed twice in PBS, DNase I was added and the samples were digested for 45 min at 37°C. Again, cells were washed, but this time they were washed in PBS-T. DNA was labeled with Alexa Fluor-647 anti-BrdU (catalog no. 560209; BD Pharmingen) and resuspended in propidium iodide (PI)-RNase staining buffer (BD Biosciences). A minimum of 105 cells were analyzed using FACSCanto and FlowJo software.
For the detection of apoptotic cells (see Fig. 5C), SDLCL cells were again grown as mixed populations of GFP-positive and GFP-negative cells after transduction with lentiviral vectors as described above. While in exponential growth phase, cells were pelleted and stained with allophycocyanin (APC)-annexin V (BD Biosciences) in binding buffer (0.01 M HEPES, pH 7.4, 0.14 M NaCl, 2.5 mM CaCl2). GFP-positive cells were analyzed for annexin V positivity using a FACSCanto flow cytometer and FlowJo software. In the inducible system, sorted GFP-positive cells were stained with both annexin V and 7-aminoactinomycin D (7-AAD; BD Biosciences) by costaining in annexin V binding buffer per the manufacturer's protocol 20 h and 44 h after the addition of doxycycline.

RNA preparation and qRT-PCR.

For analysis of miRNA expression by stem-loop quantitative RT-PCR (qRT-PCR), total RNA was prepared with TRIzol reagent according to the manufacturer's instructions and 10 ng of total RNA per reverse transcription reaction was used. Primers for RT and probes for quantitative PCR (TaqMan) were ordered from Applied Biosystems by Life Technologies and used as directed. Samples were normalized to miR-16 and RNU48 (assay identification no. 391 and 1006, respectively; Applied Biosystems). The absolute quantity of miR-155 was calculated using a standard curve of known quantities of an miR-155 synthetic RNA oligonucleotide (IDT) with the sequence 5′-rUrUrArArUrGrCrUrArArUrCrGrUrGrArUrArGrGrGrG-3′ (“r” refers to ribonucleotides).
For relative quantitation of LMP1 and EBNA2 mRNA expression levels in LCLd1/GFP and LCLd1/s155 cells, the SYBR green method was used. RNA was isolated from freshly sorted GFP-positive LCLd1/GFP and LCLd1/s155 cells by using TRIzol reagent. Equal amounts of total RNA from each sample were digested with DNase I (NEB) and reverse transcribed using the random primers supplied with the High Capacity reverse transcriptase kit (Applied Biosystems). Primers for quantitative PCR were as previously reported (3). Threshold cycle (CT) values were normalized to RNU48 mRNA levels and then expressed as values relative to the background levels of LMP1 and EBNA2 mRNA present in 293T cells, which are negative for EBV.

Cell sorting, competitive growth experiments, and cell counting.

Cells transduced with lentiviral (pLCE) sponge-containing particles—GFP, s155, and sCXCR4—were grown in R10 medium (RPMI 1640 medium with 10% FBS and 50 μg/ml of gentamicin) at 37°C for 3 days before being sorted for the same mean fluorescence intensities (MFI) of GFP on a FACSDiva flow cytometer. Cells transduced with either pMSCV- or pTRIPZ-based vectors were selected with puromycin for 1 week before being sorted for similarly high levels of GFP intensity. For pTRIPZ-transduced cells, doxycycline was added 2 days prior to sorting. Due to the approximate 5% error rate in the sorting process, cells after sorting were expected to be a mixture of ∼95% GFP-positive cells and ∼5% GFP-negative cells. After sorting, cells were resuspended in media at 2 × 105 cells/ml (except for IBL-1 cells, which were resuspended at 6 × 104 cells/ml). At the specified time points postsorting, 100 μl of cells was combined with a known quantity of AccuCount blank particles (Spherotech), 5.1 μm, and analyzed on a FACSCanto instrument (Becton Dickinson). The number of cells was calculated by normalizing to a fixed number of beads added to each sample.

RESULTS

miR-155 is the most abundant miRNA in LCLs.

Previously, it has been reported that EBV infection of resting primary human B cells results in the induction of several cellular miRNAs, including miR-155. It has also been reported that EBV-infected B cells exhibiting the viral latency III gene expression program, including LCLs and DLBCLs, express high levels of miR-155, while B cells undergoing EBV latency I, including primary EBV-positive BL tumors, express very low levels of miR-155 (6, 15, 21, 23, 26, 32, 37). To confirm the upregulation of miR-155 in the context of type III latency, we infected primary B cells with EBV and then analyzed the level of expression of miR-155 over time by using qRT-PCR (Fig. 1A). As expected, resting primary B cells, as well as the EBV-negative B-cell line BJAB and the non-B-cell line 293T, expressed low to undetectable levels of miR-155. However, infection of resting B cells with EBV caused miR-155 expression to increase throughout the 13-day time course of this experiment; at the end of the time course, miR-155 expression had been induced by >20-fold (Fig. 1A). Analysis of the two newly established LCLs EF3D and SDLCL, DLBCL cell line IBL-1 (28), EBV latency III BL cell lines Mutu III, Namalwa, and Raji, and EBV latency I BL cell line Mutu I revealed high-level expression of miR-155 in EF3D and SDLCL, undetectable levels of miR-155 in Mutu I, intermediate miR-155 expression in IBL-1, and low-level miR-155 expression in all the other B-cell lines analyzed (Fig. 1B). These data are consistent with previous reports (6, 15, 21, 23, 26, 32, 37) demonstrating that miR-155 is expressed at readily detectable levels in B cells undergoing EBV type III latency but only at very low levels in B cells undergoing EBV latency I.
We next used small-RNA deep sequencing, performed as described previously (46, 48), to analyze the total pattern of miRNA expression in the LCL EF3D. This analysis revealed that miR-155 is, in fact, the single most highly expressed miRNA in EF3D, contributing 799,949 reads (∼21%) to the total annotatable miRNA pool of 3,784,571 reads (Table 1). This analysis also identified miR-21, miR-24, and miR-146a, previously reported to also be induced upon EBV infection of B cells (6), as highly expressed cellular miRNAs, comprising ∼17%, ∼2%, and ∼2% of the total miRNA pool, respectively. In addition, we also detected the expression of eight EBV-encoded miRNAs, of which miR-BHRF1-1 was the most prominent (Table 1). Our inability to detect the other 17 miRNAs encoded by EBV results from the fact that the B95-8 EBV isolate used to generate the EF3D LCL bears a deletion that removes these miRNAs from the viral genome (5). In the EF3D LCL, all of the viral miRNAs together comprised ∼10% of the total cellular miRNA pool, i.e., less than half the level contributed by miR-155 alone.

miR-155 is critical for the growth of EBV-infected LCLs and DLBCLs.

To address whether miR-155 plays a significant role in the maintenance of EBV-infected B cells, we suppressed miR-155 activity in two recently generated LCLs (EF3D and SDLCL), in the EBV-positive BL cell lines Raji and Namalwa, and in the EBV-negative BL cell line BJAB. miR-155 activity was inhibited by transduction of these cell lines with a GFP-based lentiviral miRNA sponge specific for miR-155 (s155). We also separately transduced a previously described (17) control lentiviral miRNA sponge vector (sCXCR4), which expresses a sponge specific for an artificial small interfering RNA that can inhibit CXCR4 mRNA expression. The second control vector represents the parental lentiviral vector expressing only GFP. miRNA sponges represent a now well-established technique (4, 12, 17, 49) for specifically and stably blocking the activity of an miRNA through overexpression of an mRNA containing the gfp indicator gene linked to multiple (in this case, nine) copies of an incompletely complementary artificial target site for the miRNA of interest. Sponge-expressing cells can be sorted using flow cytometry for GFP-positive cells (17). Following transduction with each sponge and control construct, we sorted the brightest ∼20% GFP-positive cells and then monitored their subsequent growth by FACS for GFP-positive cells.
The growth of the sorted s155-expressing SDLCL and EF3D LCLs was severely attenuated compared to growth of GFP-positive cells transduced with the vectors expressing only GFP and the control sponge sCXCR4 (Fig. 2A and B, respectively). In contrast, the miR-155 sponge had no detectable effect on the growth of the EBV-negative BL cell line BJAB and the EBV-positive BL cell lines Raji and Namalwa (Fig. 2D, E, and F, respectively), even though all three of these cell lines do express detectable levels of miR-155 (Fig. 1B).
We wished to extend this analysis to the DLBCL cell line IBL-1, which is naturally infected with wild-type EBV (28), but we found that the lentiviral pLCE vector was unable to effectively transduce this cell line (data not shown). To circumvent this problem, we recloned the gfp gene, with and without the flanking miR-155 sponge, into a retroviral vector based on MSCV. Transduction of IBL-1 cells with the MSCV/s155 vector resulted in potent inhibition of the growth of GFP-positive cells, while GFP-positive IBL-1 cells that had been transduced with the control GFP expression vector replicated normally (Fig. 2C). We therefore conclude that the DLBCL cell line IBL-1 is similar to LCLs in requiring miR-155 for cell growth.
The most direct way to demonstrate miRNA function is to challenge cells with a vector in which an indicator gene has been linked to artificial target sites for that miRNA inserted into the 3′ untranslated region (16, 54). We have previously described a lentiviral indicator vector specific for miR-155 (17), and we were anxious to confirm the ability of the miR-155 sponges used in the experiments corresponding to Fig. 2 to inhibit miR-155 function. Unfortunately, the strong inhibitory effect of the miR-155 sponge on the growth and viability of the SDLCL, EF3D, and IBL-1 cells (Fig. 2A, B, and C, respectively) made this technically very difficult in these cells (this problem is addressed below). However, we were able to address whether the miR-155 sponge was active in the BJAB, Raji, and Namalwa cell lines, which remained able to replicate after transduction with the miR-155 sponge (Fig. 2D, E, and F, respectively). As shown in Fig. 3, Namalwa cells, which naturally express both miR-155 and the EBV miRNA miR-BHRF1-1, showed significant inhibition of a lentiviral indicator construct carrying the Renilla luciferase (RLuc) gene linked to artificial target sites for either miR-155 or miR-BHRF1-1 compared to 293T cells, which express neither miR-155 nor miR-BHRF1-1. As previously described (16, 17), this miRNA indicator assay is normalized by transducing cells simultaneously with both an RLuc-based lentiviral vector, containing the artificial miRNA target sites, and a firefly luciferase (FLuc)-based lentiviral vector lacking these target sites. Importantly, Namalwa cells transduced with the lentiviral miR-155 sponge expression vector failed to repress expression of the RLuc gene linked to miR-155 target sites but fully retained the ability to repress indicator vectors containing miR-BHRF1-1 target sites (Fig. 3) or target sites for miR-21 or miR-BHRF1-3 (data not shown). These data therefore argue that the miR-155 sponge is indeed able to effectively inhibit miR-155 function after transduction into the B-cell lines analyzed in Fig. 2.
To further analyze the effect of loss of miR-155 function on LCL growth in culture, we next quantified the numbers of GFP-positive and GFP-negative cells over time in cultures of the LCL EF3D that had been transduced with either s155 or GFP. As shown in Fig. 4, the s155 and GFP cells showed similar patterns of GFP expression immediately after sorting. For both samples, a small number of GFP-negative cells was present in the pool of collected cells (<5%). Six days after sorting, the number of GFP-positive cells in the s155 sample had markedly decreased, while the control GFP sample was essentially unchanged (Fig. 4, central panel). Finally, by day 25, nearly all cells present in the s155-transduced culture were GFP negative, while the control GFP-transduced cells maintained their GFP-positive phenotype. These data lend further support to the concept that LCLs lacking miR-155 function are at a severe disadvantage relative to miR-155-expressing LCLs in terms of their growth in culture. Interestingly, preliminary experiments performed in parallel using an miR-21-specific sponge failed to show any significant inhibitory effect on LCL outgrowth (data not shown), even though miR-21 is expressed at high levels in LCLs (Table 1) and has previously been associated with tumorigenesis (1, 29, 34).

miR-155 promotes cell cycle progression and inhibits apoptosis in LCLs.

To further define the basis for the observed growth deficiency of LCLs lacking miR-155, we next analyzed the cell cycle distributions of s155- and GFP-expressing LCLs. As shown in Fig. 5A and B, we observed a substantial diminution of the ability of s155-transduced cells to enter S phase relative to the abilities of the GFP and sCXCR4 controls. Consistently, the steady-state percentage of G1 cells among s155-transduced LCLs was increased relative to the percentage among controls. We further observed that the percentage of apoptotic cells, as assessed by annexin V staining, was markedly increased in miR155-deficient LCLs relative to the value for controls (Fig. 5C). A reduced ability to exit the G1 phase of the cell cycle, as well as an increased propensity to undergo apoptosis, likely explains the poor growth potential of LCLs lacking miR-155 function (Fig. 2 and 4).

Inhibition of miR-155 function by using an inducible lentiviral sponge expression vector.

A difficulty that we consistently encountered using the pLCE/s155 vector is that this vector expresses the miR-155 sponge constitutively. As loss of miR-155 expression is clearly very deleterious for LCL growth and viability (Fig. 2, 4, and 5), it is difficult to collect sufficient numbers of cells that express the miR-155 sponge for subsequent analysis. To overcome this problem, we generated tetracycline-regulatable lentiviral control and miR-155 sponge expression vectors and used these to transduce a third newly generated LCL called LCLd1. This allowed us to expand the cells after lentiviral transduction and to then induce expression of the miR-155 sponge—and the linked gfp gene—by the addition of doxycycline to the culture media prior to sorting by FACS.
As shown in Fig. 6A, this second approach again resulted in a population of GFP-positive, s155-positive cells that had lost the ability to replicate in culture, while the control GFP-positive LCLs grew normally. Analysis of miR-155 function, using an FLuc-based miR-155 indicator construct analogous to that used in the experiments corresponding to Fig. 3, revealed that the s155 sponge is able to block most, but apparently not all, miR-155 function in the transduced LCLd1 cells after doxycycline addition (Fig. 6B). The partial inhibition of miR-155 function seen in Fig. 6B may reflect the very high level of miR-155 expression in LCLs, compared to the Namalwa cells analyzed in Fig. 3 (Fig. 1B). Nevertheless, it is clear that any residual level of miR-155 function is not sufficient to support LCL growth in culture (Fig. 6A).
Because of published results suggesting that loss of miR-155 function can induce a drop in the EBV genome copy number in both LCLs and Raji cells (27), we next analyzed the expression levels of two key EBV genes in transduced LCLs by performing qRT-PCR for the viral mRNAs encoding LMP1 and EBNA2. As shown in Fig. 6C, we did not observe a significant drop in LMP1 or EBNA2 mRNA expression in the presence of the miR-155 sponge, thus demonstrating that the loss of cell growth seen upon inhibition of miR-155 function in the LCLd1 cells (Fig. 6A) is not due to a loss of LMP1 or EBNA2 expression. Finally, we also repeated our analysis of the effect of loss of miR-155 function in LCLs on apoptosis induction (Fig. 5C) by staining the transduced LCLd1 cells with annexin V, which binds to early apoptotic cells, as well as with 7-AAD, which stains cells that are in the late stages of apoptotic death. This experiment revealed levels of both annexin V and 7-AAD staining after culture of the TRIPZ/s155-transduced cells in doxycycline for either 20 h or 44 h that were substantially higher than the levels for control TRIPZ/GFP-transduced cells (Fig. 6D). These data further confirm our observation (Fig. 5C) that loss of miR-155 function induces apoptotic cell death in EBV-induced LCLs.

DISCUSSION

In this study, we confirm the previously reported (6, 15, 21, 27, 32) dramatic upregulation of endogenous cellular miR-155 expression upon EBV infection of resting B cells and demonstrate that this activation is critical for the growth and viability of the resultant LCLs and, hence, for EBV latency. Our interest in miR-155 was initially prompted by the observation that two other oncogenic herpesviruses, specifically the related human gammaherpesvirus KSHV and the essentially unrelated avian herpesvirus MDV, both encode functional orthologs of cellular miR-155 (18, 31, 42, 55). As EBV does not encode a viral miRNA with sequence homology to miR-155 yet shares with KSHV and MDV the ability to induce lymphomas in vivo, we hypothesized that EBV might instead have evolved the ability to induce cellular miR-155 expression. We further hypothesized that these induced cellular miR-155 transcripts might facilitate the establishment and/or maintenance of EBV latency, possibly by playing a critical role in the reprogramming of resting B cells into rapidly dividing EBV latency III LCLs. Our observation that miR-155 is essential for the growth of not only LCLs but also DLBCL cells in culture (Fig. 2, 4, and 6A), for their ability to cycle effectively from the G1 phase of the cell cycle into S phase (Fig. 5A and B), and for inhibition of apoptosis (Fig. 5C and 6D) is consistent with this hypothesis and suggests that KSHV miR-K11 and MDV miR-M4 may play an equivalent role subsequent to the in vivo transformation of infected lymphocytes by these herpesviruses.
All three of the LCLs analyzed in this study were generated using the EBV laboratory strain B95-8, which bears a deletion of 17 of the 22 viral BART miRNAs (5, 19). Although B95-8 is fully competent for transformation of primary B cells to LCLs in culture, it nevertheless seemed possible that these deleted viral miRNAs might be functionally redundant with cellular miR-155 and that we would therefore find that miR-155 function is dispensable in B cells transformed by wild-type EBV. To address this concern, we also analyzed the effect of loss of miR-155 function by using the DLBCL cell line IBL-1, which represents an AIDS-related lymphoma isolated from an HIV-1-positive patient (28), that expresses all 25 EBV miRNAs (data not shown). As shown in Fig. 2C, IBL-1 cells also lost their capacity to proliferate when the miR-155 sponge was introduced, thus clearly demonstrating that miR-155 is indeed required for growth of B cells transformed by wild-type EBV. Moreover, these data indicate that miR-155 is not only critically important for the growth of LCLs generated in the laboratory but also for the growth of the phenotypically similar DLBCLs that represent one of the most important categories of AIDS-related lymphomas (7).
The observation that miR-155 plays an essential role in maintaining the ability of LCLs and DLBCL cells to proliferate in culture (Fig. 2, 4, and 6A) builds on a considerable body of work documenting the critical role played by miR-155 in the normal expansion of B cells following antigenic stimulation (41, 44, 50) and also in the process of B-cell transformation following inappropriate ectopic overexpression of miR-155 (8, 9). A number of mRNA targets for miR-155 have been reported, by this laboratory and others (4, 10, 18, 27, 33, 35, 38, 39, 42, 43, 53), yet the mechanism(s) underlying the ability of miR-155 to promote B-cell expression remains unclear and is likely to be complex and multifactorial. Using the recently published photoactivatable ribonucleoside-enhanced cross-linking and immunoprecipitation (PAR-CLIP) technology (20), we have recently identified over 200 cellular mRNA targets (but no viral mRNA targets) directly bound by miR-155 in LCLs, and we are currently seeking to determine which of these are important for LCL maintenance in culture (data not shown).
One possible reason why loss of miR-155 activity might result in loss of LCL and DLBCL viability is the induction of lytic replication of EBV. Indeed, it has recently been proposed that the inhibition of bone morphogenetic protein (BMP) signaling by miR-155 stabilizes EBV latency (53). However, we did not observe any evidence for activation of EBV lytic replication in the LCLs lacking miR-155 function by FACS analysis of gp350-stained s155-expressing cells (data not shown). Moreover, and similar to results previously reported (27), we observed a slight (<2-fold) drop in viral genomic DNA copy number upon inhibition of miR-155 function, rather than the increase in genomic DNA predicted if lytic EBV replication had indeed commenced.
A final interesting observation is that while loss of miR-155 function blocked LCL and DLBCL replication, it had no significant effects on the growth of the established latency III BL cell lines Raji and Namalwa or on the EBV-negative BL cell line BJAB (Fig. 2), even though these cell lines do express miR-155, albeit at reduced levels compared to those of both LCLs and the IBL-1 cell line (Fig. 1B). The most likely explanation for this observation is that these cells have accumulated other mutations, including the c-myc translocation characteristic of BL cell lines, that render miR-155 dispensable. Indeed, EBV-positive primary BL tumors are in latency I and have been reported to express little or no miR-155 (6, 23), a result confirmed with the use of the latency I BL cell line Mutu I (Fig. 1B), so miR-155 is clearly not required for growth of BL cells. However, our data indicate that miR-155 is essential not only in newly generated LCLs but also in the phenotypically similar DLBCLs (Fig. 2, 4, and 6). These data imply that miR-155 is also likely to play a critical role in the rapidly growing EBV-positive B cells characteristic of posttransplant lymphoproliferative disorder patients (13, 24). This suggests that antisense reagents comparable to those recently shown to inhibit hepatitis C virus replication in vivo by blocking the function of cellular miR-122 (25) are worth investigating as a new approach to the treatment of both DLBCL and posttransplant lymphoproliferative disorder.
FIG. 1.
FIG. 1. Cellular miR-155 is highly induced following EBV infection of resting B cells. (A) Expression of miR-155 at the indicated time points (hours) postinfection with EBV as measured by qRT-PCR. Time zero was immediately following EBV infection. miR-155 levels in 293T and BJAB cells are shown for comparison. This graph presents the kinetics of miR-155 expression during the generation of the newly established LCL line SDLCL. Similar kinetics of miR-155 induction were observed between the primary B-cell state and LCLs by miRNA microarray analysis (data not shown). (B) Absolute quantitation of miR-155 in primary B cells and various EBV-infected B-cell lines. Results shown represent the averages of three independent qRT-PCRs, each performed in duplicate, with standard deviations (SD) indicated.
FIG. 2.
FIG. 2. miR-155 is required for growth of LCLs and DLBCLs but not EBV-positive or EBV-negative BL cells. Growth curves of GFP-positive cells expressing control sponges (black diamonds, GFP; gray triangles, sCXCR4) or a sponge specific for miR-155 (red squares, s155). GFP-positive cells (y axis) are plotted against days postsort (x axis), as determined by FACS. (A) SDLCL. (B) EF3D. (C) IBL-1. (D) BJAB (EBV negative). (E) Raji. (F) Namalwa. Graphs for panels C, D, E, and F are representative of two independent experiments, while the growth curves shown in panels A and B were determined three times. Note that the IBL-1 cells analyzed here were transduced with an MSCV-based retroviral sponge expression vector, while all other cells were transduced with lentiviral sponge expression vectors.
FIG. 3.
FIG. 3. Selective inhibition of miR-155 function in Namalwa cells transduced with an miR-155-specific sponge. Namalwa cells transduced with the control pLCE/GFP lentiviral vector or with the pLCE/s155 sponge vector were sorted for high GFP expression by FACS and then transduced with indicator vectors measuring miR-155 or miR-BHRF1-1 function. miR-BHRF1-1 activity was measured as a control for sponge specificity. 293T cells, which express neither miR-155 (Fig. 1A) nor miR-BHRF1-1, were used as a control. rel., relative.
FIG. 4.
FIG. 4. A miR-155-specific sponge strongly inhibits LCL growth in culture. The LCL SDLCL was transduced with a lentiviral GFP vector expressing a linked miR-155-specific sponge (blue) or a control lentiviral GFP expression vector (red) and allowed to grow for 6 days prior to being sorted by FACS. Levels of GFP-positive cells relative to GFP-negative cells were measured on a FACSCanto instrument immediately postsort (left panel), 6 days postsort (middle panel), and 25 days postsort (right panel). Graphs were generated using FlowJo software and are representative of three independent experiments.
FIG. 5.
FIG. 5. SDLCL cells knocked down for miR-155 function arrest in G1 and undergo increased apoptosis. (A) Cell cycle analysis of SDLCL cells expressing an miR-155-specific sponge (s155; center panel) or expressing control vectors (GFP and sCXCR4 [left and right panels, respectively]). Results were gated for the GFP-positive population, and this was done to remove sub-G1 cells as well. Numbers of propidium iodide (PI)-stained cells are plotted on the x axis, and the level of BrdU incorporation is on the y axis. Gates superimposed on each graph measure cells in the G1 (bottom left), S (top), and G2 (bottom right) phases of the cell cycle. This FACS plot is representative of three independent experiments that are quantified in panel B. (B) Cell cycle analysis of LCLs (EF3D and SDLCL), as described for panel A, but normalized relative to the GFP control sponge; results are averages from three independent experiments. Error bars represent SD. (C) Annexin V staining of SDLCL cells transduced as described above and gated on GFP-positive cells. The percentages of annexin V-positive cells shown represent averages from three independent experiments. Error bars indicate SD.
FIG. 6.
FIG. 6. Analysis of LCLs expressing a tetracycline-inducible miR-155 sponge. The corresponding experiment was performed as described for Fig. 2, except that the LCLd1 cell line was transduced either with the tetracycline-regulatable GFP expression vector pTRIPZ/GFP or with a derivative (pTRIPZ/s155) bearing the miR-155 sponge in the 3′ untranslated region (UTR) of the gfp gene. After transduction and selection for puromycin resistance, GFP expression was induced by incubation in doxycycline followed by FACS for GFP-positive cells. (A) Growth of GFP-positive LCLd1 cells transduced with either the parental TRIPZ/GFP lentiviral vector or the TRIPZ/s155 derivative. (B) Analysis of miR-155 function in doxycycline-treated LCLd1 cells transduced either with TRIPZ/GFP or with TRIPZ/s155 and then sorted for GFP expression. This experiment was performed as described for Fig. 3 and was normalized to EBV-negative cells. Results of a representative experiment reflective of two independent experiments are shown. (C) LCLd1 cells that had been transduced with TRIPZ/GFP or TRIPZ/s155 were induced with doxycycline, sorted for GFP expression by FACS, and then analyzed for LMP1 or EBNA2 mRNA expression by qRT-PCR. (D) LCLd1 cells were transduced with TRIPZ/GFP or TRIPZ/s155, expanded, and then induced by treatment with doxycycline for the indicated 20-h or 44-h time period. At this point, GFP-positive cells were analyzed for annexin V and 7-AAD expression by FACS.
TABLE 1.
TABLE 1. Summary of the miRNAs detected by deep sequencing of small RNAs expressed in the LCL EF3Da
Human pre-miRNAsb No. of readsb    
  Total 5p 3p
Cellular pre-miRNAs      
    miR-155 799,949 799,732 217
    let-7 family 747,069 747,052 17
    miR-21 655,329 654,613 716
    miR-142 203,248 7,530 195,718
    miR-103 192,754 185 192,569
    miR-320 family 134,590 1 134,589
    miR-24 93,318 21 93,297
    miR-146a 83,123 83,122 1
    miR-148a 79,124 977 78,147
    miR-29a 63,763 1,054 62,709
EBV pre-miRNAs      
    miR-BHRF1-1 152,629 151,745 884
    miR-BART1 66,816 57,423 9,393
    miR-BART15 60,893 108 60,785
    miR-BART4 29,311 27,867 1,444
    miR-BART3 18,981 10,830 8,151
    miR-BART2 17,094 17,047 47
    miR-BHRF1-2 8,000 2,577 5,423
    miR-BHRF1-3 6,641 6,587 54
a
The table presents the 10 most highly expressed cellular miRNAs detected in EF3D LCLs, as well as all eight detected EBV miRNAs, in order of expression level. For each miRNA, the total number of reads is presented for each pre-miRNA, and this number is then subdivided into reads derived from the 5′ arm or 3′ arm of the pre-miRNA hairpin intermediate. We analyzed a total of 3,784,571 assignable miRNA reads, derived from 402 cellular or viral pre-miRNA species, and the 18 presented miRNAs comprise 3,412,632 (∼90%) of this total.
b
5p, 5′ arm; 3p, 3′ arm.

Acknowledgments

We thank E. Cesarman for DLBCL cell line IBL-1. All flow cytometry was done through the Comprehensive Cancer Center's Shared Resource, and we thank Lynn Martinek, Beth Harvat, Nancy Martin, and Mike Cook for their assistance with cell sorting and analyses.
This research was supported by National Institutes of Health grants R01-AI067968 (to B.R.C.) and K99-CA137860 (to E.G.) and a joint NIH award to B.R.C. and M.A.L. (P30-AI045008) for collaborations in the study of HIV-associated malignancies. S.D.L. was supported by NIH training grant T32-AI007392, and R.L.S. was supported by T32-CA0009111. Additional research support to M.A.L. was provided by pilot grants from the Duke Comprehensive Cancer Center and the Stewart Trust.

REFERENCES

1.
Asangani, I. A., S. A. Rasheed, D. A. Nikolova, J. H. Leupold, N. H. Colburn, S. Post, and H. Allgayer. 2008. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene27:2128-2136.
2.
Bartel, D. P. 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell116:281-297.
3.
Bell, A. I., K. Groves, G. L. Kelly, D. Croom-Carter, E. Hui, A. T. Chan, and A. B. Rickinson. 2006. Analysis of Epstein-Barr virus latent gene expression in endemic Burkitt's lymphoma and nasopharyngeal carcinoma tumour cells by using quantitative real-time PCR assays. J. Gen. Virol.87:2885-2890.
4.
Bolisetty, M. T., G. Dy, W. Tam, and K. L. Beemon. 2009. Reticuloendotheliosis virus strain T induces miR-155, which targets JARID2 and promotes cell survival. J. Virol.83:12009-12017.
5.
Cai, X., A. Schäfer, S. Lu, J. P. Bilello, R. C. Desrosiers, R. Edwards, N. Raab-Traub, and B. R. Cullen. 2006. Epstein-Barr virus microRNAs are evolutionarily conserved and differentially expressed. PLoS Pathog.2:e23.
6.
Cameron, J. E., C. Fewell, Q. Yin, J. McBride, X. Wang, Z. Lin, and E. K. Flemington. 2008. Epstein-Barr virus growth/latency III program alters cellular microRNA expression. Virology382:257-266.
7.
Carbone, A., E. Cesarman, M. Spina, A. Gloghini, and T. F. Schulz. 2009. HIV-associated lymphomas and gamma-herpesviruses. Blood113:1213-1224.
8.
Clurman, B. E., and W. S. Hayward. 1989. Multiple proto-oncogene activations in avian leukosis virus-induced lymphomas: evidence for stage-specific events. Mol. Cell. Biol.9:2657-2664.
9.
Costinean, S., N. Zanesi, Y. Pekarsky, E. Tili, S. Volinia, N. Heerema, and C. M. Croce. 2006. Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proc. Natl. Acad. Sci. U. S. A.103:7024-7029.
10.
Dorsett, Y., K. M. McBride, M. Jankovic, A. Gazumyan, T. H. Thai, D. F. Robbiani, M. Di Virgilio, B. R. San-Martin, G. Heidkamp, T. A. Schwickert, T. Eisenreich, K. Rajewsky, and M. C. Nussenzweig. 2008. MicroRNA-155 suppresses activation-induced cytidine deaminase-mediated Myc-Igh translocation. Immunity28:630-638.
11.
Dull, T., R. Zufferey, M. Kelly, R. J. Mandel, M. Nguyen, D. Trono, and L. Naldini. 1998. A third-generation lentivirus vector with a conditional packaging system. J. Virol.72:8463-8471.
12.
Ebert, M. S., J. R. Neilson, and P. A. Sharp. 2007. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat. Methods4:721-726.
13.
Eis, P. S., W. Tam, L. Sun, A. Chadburn, Z. Li, M. F. Gomez, E. Lund, and J. E. Dahlberg. 2005. Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc. Natl. Acad. Sci. U. S. A.102:3627-3632.
14.
Friedländer, M. R., W. Chen, C. Adamidi, J. Maaskola, R. Einspanier, S. Knespel, and N. Rajewsky. 2008. Discovering microRNAs from deep sequencing data using miRDeep. Nat. Biotechnol.26:407-415.
15.
Gatto, G., A. Rossi, D. Rossi, S. Kroening, S. Bonatti, and M. Mallardo. 2008. Epstein-Barr virus latent membrane protein 1 trans-activates miR-155 transcription through the NF-kappaB pathway. Nucleic Acids Res.36:6608-6619.
16.
Gottwein, E., X. Cai, and B. R. Cullen. 2006. A novel assay for viral microRNA function identifies a single nucleotide polymorphism that affects Drosha processing. J. Virol.80:5321-5326.
17.
Gottwein, E., and B. R. Cullen. 2010. A human herpesvirus microRNA inhibits p21 expression and attenuates p21-mediated cell cycle arrest. J. Virol.84:5229-5237.
18.
Gottwein, E., N. Mukherjee, C. Sachse, C. Frenzel, W. H. Majoros, J.-T. A. Chi, R. Braich, M. Manoharan, J. Soutschek, U. Ohler, and B. R. Cullen. 2007. A viral microRNA functions as an ortholog of cellular miR-155. Nature450:1096-1099.
19.
Grundhoff, A., C. S. Sullivan, and D. Ganem. 2006. A combined computational and microarray-based approach identifies novel microRNAs encoded by human gamma-herpesviruses. RNA12:733-750.
20.
Hafner, M., M. Landthaler, L. Burger, M. Khorshid, J. Hausser, P. Berninger, A. Rothballer, M. Ascano, Jr., A. C. Jungkamp, M. Munschauer, A. Ulrich, G. S. Wardle, S. Dewell, M. Zavolan, and T. Tuschl. 2010. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell141:129-141.
21.
Jiang, J., E. J. Lee, and T. D. Schmittgen. 2006. Increased expression of microRNA-155 in Epstein-Barr virus transformed lymphoblastoid cell lines. Genes Chromosomes Cancer45:103-106.
22.
Kieff, E., and A. Rickinson. 2007. Epstein-Barr virus and its replication, p. 2603-2654. In D. M. Knipe and P. M. Howley (ed.), Fields virology, 5th ed., vol. 2. Williams & Wilkins, Philadelphia, PA.
23.
Kluiver, J., E. Haralambieva, D. de Jong, T. Blokzijl, S. Jacobs, B. J. Kroesen, S. Poppema, and A. van den Berg. 2006. Lack of BIC and microRNA miR-155 expression in primary cases of Burkitt lymphoma. Genes Chromosomes Cancer45:147-153.
24.
Kluiver, J., S. Poppema, D. de Jong, T. Blokzijl, G. Harms, S. Jacobs, B. J. Kroesen, and A. van den Berg. 2005. BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J. Pathol.207:243-249.
25.
Lanford, R. E., E. S. Hildebrandt-Eriksen, A. Petri, R. Persson, M. Lindow, M. E. Munk, S. Kauppinen, and H. Orum. 2010. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science327:198-201.
26.
Lawrie, C. H., S. Soneji, T. Marafioti, C. D. Cooper, S. Palazzo, J. C. Paterson, H. Cattan, T. Enver, R. Mager, J. Boultwood, J. S. Wainscoat, and C. S. Hatton. 2007. MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int. J. Cancer121:1156-1161.
27.
Lu, F., A. Weidmer, C. G. Liu, S. Volinia, C. M. Croce, and P. M. Lieberman. 2008. Epstein-Barr virus-induced miR-155 attenuates NF-kappaB signaling and stabilizes latent virus persistence. J. Virol.82:10436-10443.
28.
Lu, P., C. Yang, I. Guasparri, W. Harrington, Y. L. Wang, and E. Cesarman. 2009. Early events of B-cell receptor signaling are not essential for the proliferation and viability of AIDS-related lymphoma. Leukemia23:807-810.
29.
Lu, Z., M. Liu, V. Stribinskis, C. M. Klinge, K. S. Ramos, N. H. Colburn, and Y. Li. 2008. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene27:4373-4379.
30.
Malim, M. H., J. Hauber, R. Fenrick, and B. R. Cullen. 1988. Immunodeficiency virus rev trans-activator modulates the expression of the viral regulatory genes. Nature335:181-183.
31.
Morgan, R., A. Anderson, E. Bernberg, S. Kamboj, E. Huang, G. Lagasse, G. Isaacs, M. Parcells, B. C. Meyers, P. J. Green, and J. Burnside. 2008. Sequence conservation and differential expression of Marek's disease virus microRNAs. J. Virol.82:12213-12220.
32.
Mrázek, J., S. B. Kreutmayer, F. A. Grasser, N. Polacek, and A. Huttenhofer. 2007. Subtractive hybridization identifies novel differentially expressed ncRNA species in EBV-infected human B cells. Nucleic Acids Res.35:e73.
33.
O'Connell, R. M., A. A. Chaudhuri, D. S. Rao, and D. Baltimore. 2009. Inositol phosphatase SHIP1 is a primary target of miR-155. Proc. Natl. Acad. Sci. U. S. A.106:7113-7118.
34.
Papagiannakopoulos, T., A. Shapiro, and K. S. Kosik. 2008. MicroRNA-21 targets a network of key tumor-suppressive pathways in glioblastoma cells. Cancer Res.68:8164-8172.
35.
Pedersen, I. M., D. Otero, E. Kao, A. V. Miletic, C. Hother, E. Ralfkiaer, R. C. Rickert, K. Gronbaek, and M. David. 2009. Onco-miR-155 targets SHIP1 to promote TNFa-dependent growth of B cell lymphomas. EMBO Mol. Med.1:288-295.
36.
Pfeffer, S., M. Zavolan, F. A. Grasser, M. Chien, J. J. Russo, J. Ju, B. John, A. J. Enright, D. Marks, C. Sander, and T. Tuschl. 2004. Identification of virus-encoded microRNAs. Science304:734-736.
37.
Rahadiani, N., T. Takakuwa, K. Tresnasari, E. Morii, and K. Aozasa. 2008. Latent membrane protein-1 of Epstein-Barr virus induces the expression of B-cell integration cluster, a precursor form of microRNA-155, in B lymphoma cell lines. Biochem. Biophys. Res. Commun.377:579-583.
38.
Rai, D., S. Karanti, I. Jung, P. L. Dahia, and R. C. Aguiar. 2008. Coordinated expression of microRNA-155 and predicted target genes in diffuse large B-cell lymphoma. Cancer Genet. Cytogenet.181:8-15.
39.
Rai, D., S. W. Kim, M. R. McKeller, P. L. Dahia, and R. C. Aguiar. 2010. Targeting of SMAD5 links microRNA-155 to the TGF-beta pathway and lymphomagenesis. Proc. Natl. Acad. Sci. U. S. A.107:3111-3116.
40.
Rickinson, A., and E. Kieff. 2007. Epstein-Barr virus, p. 2655-2700. In D. M. Knipe and P. M. Howley (ed.), Fields virology, vol. 2. Williams & Wilkins, Philadelphia, PA.
41.
Rodriguez, A., E. Vigorito, S. Clare, M. V. Warren, P. Couttet, D. R. Soond, S. van Dongen, R. J. Grocock, P. P. Das, E. A. Miska, D. Vetrie, K. Okkenhaug, A. J. Enright, G. Dougan, M. Turner, and A. Bradley. 2007. Requirement of bic/microRNA-155 for normal immune function. Science316:608-611.
42.
Skalsky, R. L., M. A. Samols, K. B. Plaisance, I. W. Boss, A. Riva, M. C. Lopez, H. V. Baker, and R. Renne. 2007. Kaposi's sarcoma-associated herpesvirus encodes an ortholog of miR-155. J. Virol.81:12836-12845.
43.
Teng, G., P. Hakimpour, P. Landgraf, A. Rice, T. Tuschl, R. Casellas, and F. N. Papavasiliou. 2008. MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity28:621-629.
44.
Thai, T. H., D. P. Calado, S. Casola, K. M. Ansel, C. Xiao, Y. Xue, A. Murphy, D. Frendewey, D. Valenzuela, J. L. Kutok, M. Schmidt-Supprian, N. Rajewsky, G. Yancopoulos, A. Rao, and K. Rajewsky. 2007. Regulation of the germinal center response by microRNA-155. Science316:604-608.
45.
Thorley-Lawson, D. A. 2005. EBV persistence and latent infection in vivo, p. 309-357. In E. S. Robertson (ed.), Epstein-Barr virus. Caister Academic Press, Wymondham, Norfolk, England.
46.
Umbach, J. L., and B. R. Cullen. 2010. In-depth analysis of Kaposi's sarcoma-associated herpesvirus microRNA expression provides insights into the mammalian microRNA-processing machinery. J. Virol.84:695-703.
47.
Umbach, J. L., and B. R. Cullen. 2009. The role of RNAi and microRNAs in animal virus replication and antiviral immunity. Genes Dev.23:1151-1164.
48.
Umbach, J. L., M. F. Kramer, I. Jurak, H. W. Karnowski, D. M. Coen, and B. R. Cullen. 2008. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature454:780-783.
49.
Valastyan, S., and R. A. Weinberg. 2009. Assaying microRNA loss-of-function phenotypes in mammalian cells: emerging tools and their potential therapeutic utility. RNA Biol.6:541-545.
50.
Vigorito, E., K. L. Perks, C. Abreu-Goodger, S. Bunting, Z. Xiang, S. Kohlhaas, P. P. Das, E. A. Miska, A. Rodriguez, A. Bradley, K. G. Smith, C. Rada, A. J. Enright, K. M. Toellner, I. C. Maclennan, and M. Turner. 2007. microRNA-155 regulates the generation of immunoglobulin class-switched plasma cells. Immunity27:847-859.
51.
Wang, M., L. P. Tan, M. K. Dijkstra, K. van Lom, J. L. Robertus, G. Harms, T. Blokzijl, K. Kooistra, B. van T'veer, M. S. Rosati, L. Visser, M. Jongen-Lavrencic, P. M. Kluin, and A. van den Berg. 2008. miRNA analysis in B-cell chronic lymphocytic leukaemia: proliferation centres characterized by low miR-150 and high BIC/miR-155 expression. J. Pathol.215:13-20.
52.
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.82:5295-5306.
53.
Yin, Q., X. Wang, C. Fewell, J. Cameron, H. Zhu, M. Baddoo, Z. Lin, and E. K. Flemington. 2010. MiR-155 inhibits bone morphogenetic protein (BMP) signaling and BMP mediated Epstein Barr virus reactivation. J. Virol.84:6318-6327.
54.
Zeng, Y., E. J. Wagner, and B. R. Cullen. 2002. Both natural and designed micro RNAs can inhibit the expression of cognate mRNAs when expressed in human cells. Mol. Cell9:1327-1333.
55.
Zhao, Y., Y. Yao, H. Xu, L. Lambeth, L. P. Smith, L. Kgosana, X. Wang, and V. Nair. 2009. A functional microRNA-155 ortholog encoded by the oncogenic Marek's disease virus. J. Virol.83:489-492.
56.
Zhu, J. Y., T. Pfuhl, N. Motsch, S. Barth, J. Nicholls, F. Grasser, and G. Meister. 2009. Identification of novel Epstein-Barr virus microRNA genes from nasopharyngeal carcinomas. J. Virol.83:3333-3341.

Information & Contributors

Information

Published In

cover image Journal of Virology
Journal of Virology
Volume 84Number 2215 November 2010
Pages: 11670 - 11678
PubMed: 20844043

History

Received: 10 June 2010
Accepted: 31 August 2010
Published online: 15 November 2010

Permissions

Request permissions for this article.

Contributors

Authors

Sarah D. Linnstaedt
Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, North Carolina 27710
Eva Gottwein
Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, North Carolina 27710
Rebecca L. Skalsky
Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, North Carolina 27710
Micah A. Luftig
Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, North Carolina 27710
Bryan R. Cullen [email protected]
Department of Molecular Genetics and Microbiology and Center for Virology, Duke University Medical Center, Durham, North Carolina 27710

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy