SUMMARY
Chromatin accessibility fundamentally governs gene expression and biological response programs that can be manipulated by pathogens. Here we capture dynamic chromatin landscapes of individual B cells during Epstein-Barr virus (EBV) infection. EBV+ cells that exhibit arrest via antiviral sensing and proliferation-linked DNA damage experience global accessibility reduction. Proliferative EBV+ cells develop expression-linked architectures and motif accessibility profiles resembling in vivo germinal center (GC) phenotypes. Remarkably, EBV elicits dark zone (DZ), light zone (LZ), and post-GC B cell chromatin features despite BCL6 downregulation. Integration of single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq), single-cell RNA sequencing (scRNA-seq), and chromatin immunoprecipitation sequencing (ChIP-seq) data enables genome-wide cis-regulatory predictions implicating EBV nuclear antigens (EBNAs) in phenotype-specific control of GC B cell activation, survival, and immune evasion. Knockouts validate bioinformatically identified regulators (MEF2C and NFE2L2) of EBV-induced GC phenotypes and EBNA-associated loci that regulate gene expression (CD274/PD-L1). These data and methods can inform high-resolution investigations of EBV-host interactions, B cell fates, and virus-mediated lymphomagenesis.
Graphical Abstract
In brief
Epstein-Barr virus (EBV) persists within memory B cells in most adults and is associated with diverse cancers and autoimmunity. SoRelle et al. identify single-cell chromatin accessibility dynamics governing EBV infection. Successful infection substantially phenocopies activated, germinal center, and effector B cell states. Predicted targets regulating these phenotypes are validated experimentally.
INTRODUCTION
Epstein-Barr virus (EBV) is one of the most successful human pathogens because of its ability to achieve lifelong persistence in memory B cells (MBCs) by co-opting B cell immune responses.1 More than 95% of adults are EBV-seropositive and typically experience asymptomatic or mild illness.2 Notwith-standing, EBV is associated with diverse B cell malignancies including Burkitt lymphoma, post-transplantation lymphoproliferative disease (PTLD), and Hodgkin’s lymphoma,3 in addition to autoimmune disorders including systemic lupus erythematosus (SLE),4 and multiple sclerosis (MS).5,6
EBV engagement of tonsil germinal center (GC) B cell dynamics affords a route to latency within MBCs.7,8 However, infection yields many cellular fates. Many EBV+ B cells incur growth-associated replication defects and subsequent DNA damage response (DDR)-mediated cell-cycle arrest following de novo infection.9,10 Moreover, sustained infection within in vitro immortalized lymphoblastoid cell lines (LCLs)11,12 yields a heterogeneous continuum of phenotypes mimicking early activated GC B cells and differentiated plasmablasts.13 However, comprehensive understanding of gene regulatory control underlying these distinct outcomes remains unrealized.
Genome-wide sequencing techniques are well suited to address this knowledge gap. Ensemble RNA sequencing (RNA-seq) has revealed that EBV infection rapidly and profoundly reshapes host cell transcriptomes.14,15 Likewise, bulk assay for transposase-accessible chromatin sequencing (ATAC-seq) and viral transactivator chromatin immunoprecipitation sequencing (ChIP-seq) have provided insight into EBV-driven epigenetic rewiring (e.g., DNA methylation and histone modifications).16-19 Although these techniques highlight EBV-driven remodeling of host gene regulation, their population-averaged measurements are insensitive to heterogeneous cellular responses that occur asynchronously during infection. To resolve such dynamic biological complexity, single-cell approaches are required.20-24 Moreover, advances in single-cell multiomics and multimodal integration have facilitated comprehensive studies of hierarchical gene regulation.25 These include techniques for obtaining cell-matched measurements of mRNA transcripts, chromatin accessibility, DNA methylation status, and other molecular levels.26-29
We recently applied time-resolved single-cell RNA-seq (scRNA-seq) to develop an updated high-resolution model of early EBV infection.30 Here we present early infection single-cell ATAC-seq (scATAC-seq) data, integrate cell-matched gene expression and chromatin accessibility with bulk ChIP-seq data for host and viral proteins, and develop informatic workflows that elucidate phenotype-specific virus-mediated gene regulation. Collectively, this study provides detailed perspectives of host-pathogen dynamics that control the heterogeneous fates of EBV-infected B lymphocytes.
RESULTS
Time-resolved scATAC-seq of primary EBV infection
Single-cell chromatin accessibility (scATAC-seq) libraries were prepared from B cells prior to (day 0) and following de novo infection with B95-8 EBV31 at three time points (days 2, 5, and 8). After quality control (QC) filtering and feature selection to include the top 50% of differentially accessible peaks (DAPs), 97,191 cells across two biological donors were analyzed (52,271 and 44,920 cells, respectively). Cells from each time point contained 161,800 ATAC features on average, including >200,000 unique peaks (Figures 1 and S1A-S1D; Table S1). Time course analysis revealed global changes in cellular chromatin landscapes following infection, with the most extensive alterations occurring within two days (Figure 1A). We observed a time-dependent median of ~6,000 to ~12,000 feature peaks per cell and an experiment-wide average of ~25,000 transpositions per nucleus. Cellular chromatin accessibility was transiently but significantly reduced during early infection in both donors, with reductions consistently observed at days 2 and 5 relative to resting B cells (Figure 1B).
We next calculated chromatin accessibility trajectories using pseudotime methods32 (Figures 1C and S1E-S1H). Two main trajectories (branches) developed following infection, one of which had relatively higher net accessibility (EBV+-high ATAC) than the other (EBV+-low ATAC). Although EBV+-low ATAC cells occurred late in pseudotime, they were detectable by day 2. Both branches contained cells from each post-infection timepoint and recapitulated time-resolved trends (Pearson R = −0.32). This bifurcation of global chromatin states corresponded to broad changes in gene regulation following infection. Cis-linked regulatory prediction33 coupled with gene ontology analysis identified significantly enriched DAPs in EBV+-low ATAC and EBV+-high ATAC states versus uninfected B cells (Table S1). The top 2,000 ATAC sites with significantly enhanced accessibility in resting B cells versus EBV+ branches yielded predicted linkages to >1,800 genes. DAP-linked genes were significantly associated with ~200 Gene Ontology (GO) biological process terms, especially those related to mRNA metabolism, chromatin organization, gene expression, and B cell activation (Table S1). The top 2,000 significant sites with greater accessibility in EBV+-high ATAC cells versus resting cells had predicted linkages to nearly 1,500 genes. Significant GO terms for EBV+-high ATAC DAP-linked genes were related to lymphocyte activation, defense responses, cytokine production, and programmed cell death (Table S1). By contrast, EBV+-low ATAC cells only exhibited 170 genes linked to 174 significant DAPs with enhanced accessibility versus resting B cells. Only one GO term (regulation of immune process, GO:0050776) was enriched in EBV+-low ATAC cells versus resting cells. Thus, although EBV+-low and EBV+-high ATAC states exhibited reduced accessibility versus uninfected cells, only EBV+-high ATAC cells gained accessibility at loci regulating B cell activation and immune responses.
We performed multimodal integration of cell-matched scATAC-seq and scRNA-seq data using Signac.34 Clusters identified via weighted nearest neighbor analysis were mapped to early infection phenotypes defined in prior work30 (Figures 1D and S1). These phenotypes include uninfected naive (c8) and memory (c3) B cells, EBV+ cells undergoing innate antiviral sensing (c4), EBV+ cells exhibiting growth- and DNA damage-induced arrest (c7), EBV+ GC dark zone (DZ)-like hyperproliferating cells (c6), EBV+ activated precursor/early MBCs (AP-eMBCs; c0), naive-derived EBV+ activated cells (c1), EBV+ GC light zone (LZ)-like NF-κB activated cells (c2), and EBV+ differentiated plasmablasts (c5). Integration enabled cell mapping by timepoint, ATAC trajectory, and biological phenotype. EBV+-low ATAC cells with significantly reduced accessibility corresponded to unsuccessful infection (c4 and c7; Figure 1E). By contrast, uninfected (c3 and c8) and hyperproliferative (c6) cells had the most open chromatin. Hyperproliferative cells (c6) exhibited variable accessibility attributable to a composition of proliferating cells (higher accessibility) and those trending toward oncogene-induced arrest (lower accessibility). There were significantly more ATAC peaks in NF-κB activated cells (c2) relative to other activation intermediates (c0 and c1) and differentiated cells (c5). Significant cluster-resolved DAPs, cis-linked genes, and GO enrichment from DAP-linked gene predictions are provided in supplemental information (Table S2).
Because accessibility was recovered between days 5 and 8, state-resolved profiles indicated that EBV-induced chromatin closure is transient during successful infection. Thus, the EBV+-low ATAC trajectory defines cell arrest and the EBV+-high ATAC trajectory leads to immortalization. Accordingly, significantly depleted accessibility was observed in arrest states, especially c4, at loci such as the AP-1 transcriptional activator BATF (Figure 1F), which is essential for B cell activation and LCL survival.35 GO analysis of DAP-linked differentially expressed genes (DEGs) between resting, EBV+ arrest, and EBV+ success groups confirmed closure at immune cell activation loci in EBV+ arrest and identified accessibility-linked expression of latency establishment mediators (e.g., NFKBIA, RBPJ) in EBV+ success states versus resting B cells. B cell activation, antiviral response, and survival genes linked to accessibility in immortalizing but not arresting trajectories included AICDA, BCL2, CD83, IFNG, PRDM1, and TRAF2.
Linked expression and accessibility illuminate phenotype-specific regulation
We summarized gene regulation across different EBV+ fate trajectories. Nine hundred fifty-four mRNA features were derived from the top 100 markers for each early infection phenotype. One hundred seventy-seven of these DEGs were significantly linked to 476 DAPs (18.6% of tested genes with potential DAP-linked regulation; Figures 2A and S2). DAP-linked DEG analysis revealed four regulatory patterns: higher accessibility linked to higher expression (+/+), lower accessibility linked to higher expression (−/+), lower accessibility linked to lower exp ression (−/−), and higher accessibility linked to lower expression (+/−). The +/+ and −/− patterns were characteristic of accessible versus inaccessible positive regulatory loci, respectively. The less frequently observed −/+ and +/− patterns were consistent with closure or opening of ATAC sites associated with negative regulation. Genes of interest including CCR7, CXCR4, RUNX3, BACH2, JCHAIN, and PRDM1 provided examples of each pattern and their phenotype-specific variation (Figure S2). For example, seven significant expression-linked DAPs (five positive and two negative regulatory loci) were identified within 20 kB of the transcription start site (TSS) for CCR7. The BACH2 locus contained 13 significant intragenic regulatory loci including two negative sites. As for CCR7 and CXCR4, all positive regulatory loci linked to BACH2 expression exhibited reduced accessibility in EBV+ phenotypes. By contrast, RUNX3, PRDM1, and JCHAIN had more open chromatin at positive regulatory sites in EBV+ states versus resting B cells, with PRDM1 and JCHAIN specific to EBV-induced plasmablasts (c5).
Because DAP-linked DEGs included numerous B cell fate mediators, we investigated regulatory relationships in distinct trajectories (Figures 2B-2D). In the path from resting to arrested/senescent cells (c4 and c7), 23.4% of DEGs were linked to DAPs that became inaccessible after infection (i.e., [3 ∪ 8] ! [4 ∪ 7] peaks; Figure 2B). DAP-linked DEGs downregulated in this trajectory included CFLAR and NFKB2, both of which are essential in LCLs.35 Similar DAP-linked loss of basal expression from uninfected to arrested states was frequently observed for genes involved in B cell signal transduction (e.g., CD74, FYN, INPP5D, LYN, MAP4K3, PRKCE) and reflected in top GO terms (e.g., intracellular signal transduction, lymphocyte activation, regulation of BCR signaling pathway; Table S3). Accessibility loss in c7 was consistent with induction of senescence-associated heterochromatin foci (SAHF; Figures S3A and S2B).36-38 Because senescence can arise from diverse mechanisms such as innate sensing or DNA damage, we used higher resolution clustering to refine c7 subsets (7a and 7b). These subsets displayed DEGs involved in the cell cycle and antiviral sensing (Figures S3C-S3F). Different HMGB2 levels between 7a and 7b were notable, as HMGB2 mediates diverse roles in sensing,39 double-stranded break repair,40 and p53 downregulation.41 HMGB2 expression was elevated in 7b (as in hyperproliferative cells) versus 7a (similar to c4, which could precede senescence42). Proliferation markers were also lower in 7a than 7b. Thus, 7a corresponded to EBV+ cells that arrested rapidly via innate sensing, while 7b corresponded to later arrest induced by replicative stress.
NF-κB activated EBV+ cells (c2) exhibited diminished accessibility at 1,142 sites relative to resting cells (8.7% of resting cell peaks). However, c2 cells possessed 668 peaks absent in resting cells ([2] ! [3 ∪ 8]) linked to 595 genes. One hundred fifty-four of these (25.9%) were DEGs between resting and activated cells (Figure 2C), including 109 upregulated and 45 downregulated genes. Upregulated genes along this trajectory included IRF4 and CCND2, both of which are LCL dependencies.35 The IRF4/DUSP22 locus contained at least nine positive regulatory loci that were more accessible in the EBV+ NF-κB activated state versus uninfected cells. Other upregulated genes included mediators of apoptosis and tumor suppression (BCL2A1, TNFRSF8, PDCD1LG2, ST7, IQGAP2, TOPBP1, CD86); proliferation (CDCA7L, MKI67); and B cell signaling (NFKBIA, MAPK6, TNIP1, TRAF3). Collectively, (2) ! (3 ∪ 8) DAP-linked DEGs underscored the importance of host chromatin remodeling for cell activation, proliferation, and apoptotic resistance facilitating immortalization (Table S4).
We next explored DAP-linked DEGs between EBV-induced NF-κB activation (c2) and differentiation (c5; Figure 2D). Activated cells exhibited 999 peaks absent from differentiated cells ([2] ! [5] DAPs), while differentiated cells displayed only 13 exclusive peaks, corresponding to a 15% reduction in accessibility. Notably, (2) ! (5) DAPs were linked to 13.4% of all DEGs between these states identified from scRNA-seq. One example was the TNFAIP3 (A20) locus, which contained ten significant linked positive regulatory loci. Conversely, DAP-linked DEGs upregulated from c2 to c5 included plasmablast-specific transcriptional regulators (e.g., XBP1, PRDM1), translation factors, and protein export machinery (i.e., facilitators of Ig synthesis and secretion; Table S4). CD38, a marker of MBCs, was also a DAP-linked DEG with enhanced accessibility in c5 versus c2. These results demonstrate that successful infection reprograms cellular chromatin not only to support activation, proliferation, and survival but also the subsequent transition to effector states.
Decreased accessibility at EBNA binding sites silences B cell response genes in EBV+ arrest
We examined gene silencing associated with inaccessible chromatin in EBV+ arrest, particularly at expression-linked DAPs coinciding with EBV nuclear antigen (EBNA) ChIP-seq footprints (Figure 3A). We preliminarily validated reductions in ADAM28 and COBLL1 expression, which are epigenetically suppressed targets of EBNA-3C,43,44 and found that downregulation of these genes was extensively linked to chromatin closure at EBNA-3C binding sites (Figure S4). Consistent with GO results from DAPs alone, many genes that mediate B cell signal transduction and activation displayed accessibility-linked repression in EBV+ arrest. Frequently, expression of these genes was linked to multiple regulatory loci including EBNA-LP, EBNA-2, and/or EBNA-3C ChIP-seq sites (Figures 3B-3F). Expression-linked DAPs at the IL21R locus, including two EBNA-2 sites, were significantly depleted in EBV+ arrest (c4 and c7) relative to resting B cells and successful infection (Figure 3B). This finding was noteworthy, since B cell-specific IL21R signaling is essential for GC engagement in vivo.45-47 Similarly, EBV+ arrest states displayed accessibility-linked repression across the CD58 (LFA-3) locus, including an EBNA-LP site (Figure 3C). Although loss of CD58 expression facilitates escape from immune surveillance in B cell lymphoma,48 CD58 engagement in vivo with T cell-expressed CD2 is essential for actuating B cell responses to antigen.49 EBV+ arrest states also exhibited DAP-linked downregulation at EBNA ChIP-seq sites near MAPK8 and PDE4D, which encode B cell receptor (BCR) and Toll-like receptor (TLR) signaling mediators (Figures 3D and 3E).50,51 Four peaks including three expression-linked DAPs upstream of the Jun N-terminal kinase (JNK)-encoding MAPK8 locus coincided with EBNA-2 sites (Figure 3D). Accessibility-linked MAPK8 loss in EBV+ arrest states is predicted to inhibit B cell survival and transformation on the basis of the gene’s essentiality.50 The enzyme encoded by PDE4D degrades the secondary messenger cAMP, which, in activated T cells, removes a restraint to TCR signaling.52 By analogy, accessibility-linked loss of PDE4D expression in B cells (Figure 3E) may impede signal transduction and NF-κB activation downstream of viral LMP-2A, which mimics BCR signaling,53 via cAMP accumulation.54 Finally, EBNA-associated accessibility-linked expression was observed for S1PR2, which encodes a GC B cell-specific surface receptor and helps maintain GC localization and homeostasis.55 Although higher S1PR2 expression was observed in c7 and EBV+ success states, c4 cells did not exhibit accessibility-mediated S1PR2 upregulation (Figure 3F). A similar pattern was observed at the adjacent DNMT1 locus, whose product is a DNA methyltransferase that restrains viral latency.56 These findings suggest that failure of EBNAs to maintain or induce accessibility-linked expression of B cell signaling and adaptive response genes contributes to EBV+ arrest outcomes.
Downregulation of DAP-linked DEGs involved in proliferation also defined EBV+ arrest (Figures 3G-3J). CDCA7 (JPO1), a MYC target that promotes lymphoblastic transformation,57,58 was expressed in successful infection but not resting or EBV+ arrested cells (Figure 3G). CDCA7 expression was significantly linked to an upstream EBNA-3C associated DAP. Consistent with prior work dissecting EBV-induced arrest versus proliferation,10 MKI67 expression was significantly lower in arrest versus success trajectories. Attenuated MKI67 expression in c4 was linked to reduced accessibility at an upstream EBNA-LP site (Figure 3H). An EBNA-2 associated DAP linked to expression of the DNA polymerase (Pol) catalytic subunit POLA1 was closed in arrest phenotypes, implying defective DNA replication (Figure 3I). Another significant EBNA-associated DAP-linked DEG, NEK2, encodes a centrosome-associated kinase that regulates mitosis (Figure 3J). Loss of accessibility and NEK2 expression in c4 further indicated impaired proliferation.
Accessibility-mediated repression of these genes along the arrest trajectory was consistent with inhibited cell activation and proliferation, which contrasts with functional B cell adaptive responses and successful EBV infection. Collectively, these data support an antiviral role for targeted host chromatin closure as well as the necessity for viral oncoproteins to counteract this restriction mechanism to establish latency.
EBV induces GC B cell-like chromatin during successful infection despite BCL6 downregulation
Next, we investigated DAP-linked DEGs between uninfected B cells and EBV+ success phenotypes (c6: GC DZ-like; c0: AP-eMBC-like; c2: GC LZ-like; and c5: post-GC memory/plasmablast; Figure 4A). Because EBV induces GC-like expression signatures,7,30,59-61 we focused our analysis on DAP-linked DEGs that are GC B cell biomarkers and transcriptional regulators and integrated ATAC-seq data from tonsillar B cells8 and EBNA ChIP-seq data.
BCL6 is the master regulator of GC B cell responses. Although BCL6 mRNA is expressed in resting naive B cells, BCL6 protein expression in vivo depends on activation and T cell help.62 Consistent with BCL6-repressive functions of EBV microRNAs and EBNAs,63-65 rapid BCL6 loss was observed in all post-infection states (Figure 4B). BCL6 downregulation in EBV+ success (c6, c0, c2, and c5) was linked to closure of an upstream locus, which was also inaccessible in tonsillar GC DZ, LZ, and plasmablast subsets. An adjacent EBNA-LP binding site was accessible across tonsillar GC subsets but became inaccessible in EBV+ states in vitro; this locus was not significantly linked to BCL6 expression. Further comparisons of de novo EBV ATAC profiles with analogous tonsil phenotypes revealed divergence from conventional GC B cell programming. For example, the GC DZ homing receptor CXCR462,66 was downregulated upon infection as found in prior studies.67 CXCR4 repression was significantly associated with closure of multiple EBNA-associated DAPs (Figure 4C). Again, reduced accessibility at several of these sites contrasted with open chromatin in tonsillar DZ B cells.
Despite repression of BCL6 and CXCR4, chromatin profiles at many loci encoding GC biomarkers and transcriptional regulators were faithfully recapitulated during EBV infection. For example, c6 (DZ-like) and c2 (LZ-like) EBNA-associated loci near LMO2, an oncoprotein expressed in GCs and GCBDLBCL,68 closely resembled accessibility in GC DZ and LZ cells (Figure 4D). Likewise, peaks exclusive to tonsillar DZ and LZ cells near the GC hallmark AICDA69,70 were also uniquely accessible in c2 and c6 during early infection (Figure 4E). A multi-EBNA site upstream of the AICDA TSS was only accessible in LZ tonsil cells in vivo but was open in EBV-induced DZ-like (c6) and LZ-like (c2) states. The transcription factor BHLHE40, which is required for and restrains the GC reaction in vivo,71 had an upstream EBNA-associated expression-linked DAP not observed in tonsil subsets. This peak was absent in resting cells in vitro but accessible in c2 (Figure 4F).
The closest EBV-induced mimicry of GC B cell chromatin and expression was observed between c2 and LZ cells. Although expression of the GC LZ marker CD83 was downregulated in the ensemble cell population following infection, its accessibility-linked expression was conserved in c2 (Figure 4G). Although this was consistent with LMP-1 upregulation of CD83 through NF-κB signaling,72 significant linkages to EBNA-2 and EBNA-3C DAPs suggest that other latency genes may promote or maintain CD83 expression. CD86, another marker of activated LZ B cells, was likewise enriched in tonsil GCs and c2, with significant linkage to an EBNA-LP DAP (Figure 4H). EPHB1, a marker of LZ MBC precursors in vivo,73 was expressed in c2 and linked to two EBNA-associated DAPs (Figure 4I). One locus was consistently accessible in tonsil DZ, LZ, and plasmablasts, while another was a multi-EBNA binding site inaccessible in tonsils. EBV-induced EPHB1 expression was noteworthy with regard to immune evasion and viral malignancy, as EPHB1 limits TFH cell recruitment to GCs in vivo 74 and is a marker of EBV+ Reed-Sternberg cells.75
Collectively, this analysis identified extensive virus-driven alterations of host chromatin at genes critical to GC B cell dynamics. Several discrepancies, especially BCL6 downregulation, underscore the dysregulation of EBV-driven dynamics relative to in vivo GCs. However, the extent of EBV-induced GC B cell chromatin and gene expression mimicry reveals viral subversion of host adaptive responses at multiple regulatory levels.
EBV modulates accessibility at transcription factor binding motifs in a dynamic GC-like reaction
To further investigate phenotype-specific regulation, we assayed TF binding site (TFBS; i.e., motif) enrichment by state (Figure S5). We found variable motif enrichment linked to resting cells and among non-arrested EBV+ phenotypes (Figure S5A). Variable motif accessibility broadly matched gene expression with respect to antiviral response induction, cell growth, and oncogenesis (Figures S5B and S5C). Activated B cell (c2) ATAC peaks were enriched in binding sites for proto-oncogenic TFs in the REL (cREL, RELA, RELB), AP-1 (FOS, FOSB, JUNB, JUND), and EGR (EGR1–4) families. Enhanced accessibility at NF-κB TFBS in c2 was consistent with the corresponding upregulation of NF-κB targets. Likewise, differentiated cells (c5) were enriched in accessible motifs for IRF4, IRF8, and XBP1. Globally, resting B cells and the innate sensing arrest state shared the greatest motif correlation (R > 0.75) and the lowest correlation to EBV-activated and hyperproliferating cells (0.55 < R < 0.7; Figure S5D).
We examined EBV-induced reprogramming through integrated analysis of motif accessibility and expression (Figure 5). Differentially accessible motifs between uninfected naive versus MBCs were reflected within infected non-GC versus GC (DZ and LZ) phenotypes. TFBS with enhanced accessibility in EBV− MBCs and EBV+ LZ cells included motifs for JDP2, FOS/JUN AP-1, and NFE2L2. Conversely, motifs with enriched accessibility in EBV− naive B cells and EBV+ non-GC cells included those for POU2F2, BHLHE40, and MEF2C (Figures 5A and 5B). We characterized motif enrichment within EBV+ non-GC, LZ, DZ, arrest, and pre-lytic states (Figure 5C) and mapped scRNA-seq clusters onto the motif accessibility landscape (Figure 5D). Pairing this integration with pseudotime analysis resolved dynamic motif accessibility signatures for virus-induced GC entry, engagement, and exit. In addition to increased NF-κB, REL, FOS, and JUN family motif accessibility in LZ cells, we observed anticorrelated YY1 and BHLHE40 motif accessibility in the transition to the LZ state (Figure 5E).
To assay functional roles of these transcription factors in supporting GC-like transitions, we knocked out each of four TFs whose motifs were either enriched (JDP2 and NFE2L2) or depleted (BHLHE40 and MEF2C) between LZ-like (c2) and non-GC/differentiated (c5) states. We used a fluorescence-activated cell sorting (FACS)-based assay to capture dynamic transition between these states,59 where sorting and replating LCLs from the c2/GC LZ state (ICAM1hi/CD27lo) enables monitoring of their transition to the c5/differentiated (ICAM1lo/CD27hi) state (Figure 5F). CRISPR knockout (KO) of each TF was assessed by co-KO of the inert proxy CD46 76 and validated by sequencing to confirm genetic lesions (Figure S6A). Functional NFE2L2 (NRF2) KO was further validated by western blot (Figure S6B). NFE2L2 KO yielded faster accumulation of differentiated cells, consistent with its predicted role in restraining this transition (Figures 5B and 5F). In contrast, loss of functional MEF2C via MADS DNA binding domain deletion77 impeded accumulation of differentiated cells observed in the control CD46 KO (Figures 5B, 5F, and S6). We did not observe significant effects of JDP2 or BHLHE40 KO on activation and differentiation states, suggesting that these were not critical in regulating this transition in LCLs.
Post-infection phenotypes resolved by differential EBV episomal locus accessibility
We also investigated viral genome accessibility during early infection. Twenty-one viral ATAC peaks (20 of 21 in both donors) were detected, including at TSSs for essential latency genes such as the EBNAs and LMP1. Episome ATAC signals in a negligible number of B cells at day 0 (n = 3, <0.02% of cells) were attributed to pre-existing wild-type infection (Figure S7A). EBV+ activated (c2) and hyperproliferative cells (c6) had the greatest number of accessible episomal loci relative to other post-infection phenotypes. These sites included the C promoter (Cp) for EBNA1, EBNA2, and EBNA3A–3C; the LMP1 TSS; the TSS for BMRF1, a DNA Pol accessory protein; and the BHLF1 locus, which facilitates latency and immortalization (Figures S7B and S7C).78 Innate sensing (c4) and growth arrest states (c7) had the fewest cells with episomal ATAC peaks (Figure S7D).
Informatic inference of phenotype-resolved TF signatures and regulatory elements
The prevalence of DAP-linked DEGs modulated by EBV gene products led us to interrogate genome-wide phenotype TF signatures. We reasoned that ensemble-averaged ChIP-seq data from LCLs would contain TF binding and epigenetic data representing a superposition of EBV+ phenotypes at high coverage, thus maximizing chances to identify overlaps with comparatively sparse scATAC-seq data. We further extrapolated that phenotypic variation in TFBS accessibility would underlie distinct infection outcomes. Finally, empirical differential expression measurements of genes identified by cross-referencing cluster ATAC peaks with ChIP footprints of interest could be retrieved from cell-matched scRNA-seq. This tri-modal integration was expected to clarify the heterogeneity of putative regulatory relationships in early infection (Figure S8A). Thus, we employed a bioinformatic workflow to obtain ChIP-seq referenced inference of single-cell phenotypes from scATAC-seq data (“crisp-ATAC”). This process applies genomic range intersection “recipes” between scATAC-seq clusters and bulk ChIP-seq datasets in conjunction with cis-regulatory prediction (Figures S8B). During initial testing, we found that logic-gated peak intersections across multiple ATAC-seq and ChIP-seq datasets frequently exhibited “noisy” short intervals (<250 bases) due to minor offsets in called peaks across input datasets. However, crisp-ATAC consistently recovered “signal” intervals (~1,100 base median) representing DAPs associated with ChIP-seq signals from the applied recipe (Figure S8C). Most noise intervals could be suppressed by incorporating biological replicates. Specifically, DAPs between the same phenotype across replicates were dominated by noise, whereas DAPs between different phenotypes and different replicates contained noise and signal intervals. Logical gating that recovered DAPs present in one phenotype and absent in another across replicates effectively filtered noise (Figure S8D). This allowed comparisons of DAP frequencies and interval length distributions across all pairwise phenotype comparisons (Figure S8E). Thus, replicate-based interval filtering provided essential QC for all crisp-ATAC predictions.
Crisp-ATAC finds TF-linked expression signatures that vary by EBV-induced fate
We used crisp-ATAC to predict EBNA and LMP1-mediated NF-κB accessible sites at promoters, enhancers, and actively transcribed genes in each phenotype from ChIP-seq peaks for EBNAs,44,79,80 NF-κB/Rel TFs,81,82 H3K4me1, H3K4me3, H3K27ac, H3K36me3,83 and RNA Pol II84,85 (Figure S8F). Hyperproliferative (c6), activated (c2), and resting MBCs (c3) exhibited up to 3-fold more enhancers and promoters at EBNA-2 sites relative to naive B cells (c8), other activation intermediates (c0 and c1), plasmablasts (c5), arrested states (c4 and c7), and non-B cells (c9 and c10). Similar patterns were observed for EBNA-3C and EBNA-LP as well as Rel family TFBSs (Figure S8G). Enhancers, promoters, and actively transcribed genes were enriched in c2, c3, and c6 and depleted in c4 and c7, with intermediate levels in c0, c1, c5, and c8.
We assayed regulatory variation associated with key viral transcriptional co-activators, starting with innate sensing arrest (c4) and NF-κB (c2) states (Figure 6A). Peaks were extracted and gated to obtain (2) ! (4) DAPs (n = 1,873) and cis-linked gene predictions (n = 1,514; Figures 6B and 6C). The (2) ! (4) linked gene ontology was enriched for innate defense (inflammation, antimicrobial processes) and EBV-induced responses (lymphocyte activation, regulation of apoptosis; Figure 6D; Table S4). Overall, (2) ! (4) linked gene predictions included 42.5% (71/167) of known EBV super-enhancer (EBVSE) site-linked genes.18 Consequently, 41%–55% of EBNA-associated DAPs overlapped a peak for the super-enhancer-associated host TF RelA. Of 71 identified EBVSE-linked genes, 19 (27%) were linked to EBNA-LP peaks, 22 (31%) to EBNA-2 peaks, and 15 (21%) to EBNA-3C peaks. EBVSE-linked genes were enriched in EBNA-associated DAP-linked DEGs relative to size-matched random samples of the captured transcriptome (Figure 6E).
We analyzed genes of interest on the basis of EBVSE status, GO process involvement, and/or empirically demonstrated importance to EBV infection. The NF-κB activated gene and signal transducer TRAF1, whose gene product interacts with LMP-1, matched all three criteria above.86-90 We found (2) ! (4) expression-linked DAPs associated with one or more EBNA at −3, +2, and +37 kb from the TRAF1 TSS (p < 0.05, correlation Z score). Furthermore, these loci exhibited reduced accessibility in arrest (c7), activation intermediate (c1) and differentiated (c5) states versus c2 (Figure 6F).
A grouped comparison ([2 ∪ 5 ∪ 6] ! [3 ∪ 8]) identified changes associated with successful EBV-induced immortalization (Figure 7; Table S5). Despite net accessibility loss after infection, 245 genes linked to 1,824 peaks were present in the EBV+ states but not resting cells (Figures 7A-7C). One hundred sixty-six of 245 genes were linked to at least one EBNA site, and 18 genes overlapped with EBVSE targets (7.3% of predicted genes, 10.8% of known EBVSE genes). Only 31 GO terms were shared across the top 100 terms for each EBNA, accounting for 15% of unique terms (Figure 7D). We selected the (2 ∪ 5 ∪ 6) ! (3 ∪ 8) ∩ EBNA targets TNFRSF8 (CD30), CD274 (PD-L1), and PDCD1LG2 (PD-L2) based on their relevance to EBV-associated lymphomas. These included three EBNA-2 sites near the TNFRSF8 TSS (−17, −12, and +16 kb) and a multi-EBNA site −17 kb from the PDCD1LG2 TSS and +43 kb from the end of CD274 (Figure 7E). These loci were enriched for Rel sites and activating histone marks in LCLs. Cas9/RNP KOs were used to investigate the functional role of the putative enhancer located between PDCD1LG2 and CD274 (PD-L1). Relative to an adjacent control region, deletion of this enhancer led to a reduction in PD-L1 expression (Figure 7F), supporting a role for viral oncoprotein-mediated alterations to chromatin accessibility that facilitate immunoevasion.
In a final example, we evaluated activated versus differentiated EBV+ phenotype DAPs ([2] ! [5]) associated with EBNA binding to explore regulatory control delineating key phenotypes present in LCLs 13 (Figure S9A). Five hundred nineteen of 999 identified (2) ! (5) peaks intersected with at least one EBNA site, from which 247 genes were predicted. Twenty-nine of 110 (2) ! (5) ∩ EBNA-2 site-linked genes (26.3%) were DEGs in the scRNA assay, as were 34 of 115 (2) ! (5) ∩ EBNA-LP site-linked genes (29.6%) and 20 of 125 (2) ! (5) ∩ EBNA-3C site-linked genes (16.0%). Twenty genes were identified by including all three EBNAs (Figure S9B), including the EBVSE-linked GPR137B, a regulator of mTORC1 activity and autophagy.91,92 GPR137B was also a (2) ! (3 ∪ 8) DAP-linked DEG, indicating that loci regulating GPR137B expression after infection were initially inaccessible within resting cells. Two EBNA-associated expression-linked loci were found +14 and +18 kb relative to the GPR137B TSS. One site (+14 kb) coincided with EBNA-LP and EBNA-3C binding sites but did not exhibit a (2) ! (5) DAP. The second site (+18 kb) overlapped with all three EBNAs and was a DAP. Both sites intersected with Rel family TFs (Figure S9C). Other genes involved in lysosome-mediated processes including autophagy and antigen presentation regulation were identified from the (2) ! (5) comparison (TFEB and LAMP3), albeit with modestly elevated but significant expression.93-95 Other EBNA-associated DAP-linked DEGs involved in immune signaling, apoptosis, and transcription were also identified (Figures S9D and S9E). Collectively, crisp-ATAC analyses of early infection phenotypes refined the genome-wide dynamics of heterogeneous EBV-induced B cell responses.
DISCUSSION
This study provides a detailed view of chromatin accessibility landscapes that develop upon EBV infection. To facilitate high-resolution investigations of EBV-mediated regulatory control, we present annotated infection phenotypes determined from single-cell multiomics. Bioinformatic integration enabled gene- and peak-level interrogation of host and viral epigenetic signature variation between these phenotypes. The combination of resolution, multimodality, and integrative analysis yield a vividly detailed representation of the genome-wide interplay of host and virus.
Because of asynchronous parallel emergence of heterogeneous cell responses, early infection chromatin dynamics cannot be fully resolved with ensemble sequencing. Remarkably, we find transient genome-wide euchromatin-to-heterochromatin transitions (20%–40% reduced accessibility) in some post-infection trajectories. Cell-to-cell variation in chromatin accessibility following infection warrants consideration of Simpson’s paradox. That is, the total number of unique peaks across ensemble populations increases after infection (Table S1; consistent with prior studies), while the number of peaks per cell in fact decreases along several trajectories.96,97 Although scATAC-seq data sparsity likely contributes to underestimation of accessible loci, variable peak signatures and frequencies among distinct fates suggest the importance of a balance between drivers of epigenetic silencing (e.g., arrest and senescence via antiviral and damage response induction) and activation (e.g., oncogenic EBVSE formation) in determining post-infection cell fate.
In oncogene-induced senescence, DNA damage triggers SAHF formation and epigenetic silencing.37 Our lab found that EBV induces γH2AX foci via ATM/Chk2-mediated DDR signaling 9 and H3K9me3 heterochromatin induction10 in cells that arrest during viral oncoprotein-driven hyperproliferation. Another subset of EBV+ cells failed to proliferate but also exhibited DDR induction on the basis of γH2AX and pChk2.9 Similarly, here we find substantial loss of accessible chromatin and expression in cells that undergo innate sensing-mediated arrest (c4) or evade this response but arrest due to DDR activation (c7) during hyperproliferation (c6). Our data also indicate a role for ribosome biogenesis in the transition from virus-induced arrest to senescence, likely through a p53-MDM2 axis,98 which regulates EBV transformation.99 Chromatin closure in EBV-induced arrest was enriched at loci linked to B cell activation, signaling, and proliferation. Many such sites overlap with footprints for EBNAs, several of which (EBNA-1, EBNA-3C) have been shown to directly promote DNA damage through mechanisms including double-stranded break induction.100 Moreover, EBNA-LP co-localizes with γH2AX foci9 and binds to proteins within promyelocytic leukemia nuclear bodies (PML NBs) that sense DNA damage.101,102 Because ATM-mediated DDR silences genes near DSBs by impeding RNA Pol II-driven chromatin decompaction,103 it is conceivable that local damage at EBNA sites contributes to this targeted heterochromatinization in EBV-induced arrest. Although precise mechanisms are unclear, this finding suggests that DDR-induced heterochromatinization in response to EBV oncoprotein function simultaneously enacts antiviral restriction and ablates the infected cell’s adaptive response capacity.
Successful infection is distinguished by increased accessibility near key genes against the backdrop of heterochromatin induction, presumably by avoiding extensive DDR activation.9,10 Likewise, episomal peak heterogeneity across EBV+ phenotypes confirms that latency establishment depends on retained accessibility to viral genes within the repressive host milieu. In successful infection, a substantial number of accessible cellular loci have predicted cis linkages to genes enriched for regulation of apoptosis, tumor suppression, inflammation, and chromatin remodeling. We found that 10 of 87 genes (11.5%) shown to be essential for EBV-induced B cell immortalization35 had significant expression-linked DAPs. Seven of these (BATF, CDK6, CFLAR, IRF2, NFATC2IP, NFKB2, and SP110) exhibited loss of basal accessibility in EBV+ arrest relative to EBV+ success trajectories. Conversely, DAPs at three genes (CCND2, IRF4, and MDM2) were closed in resting cells and failed to open in EBV+ arrest but not EBV+ success trajectories. Thus, maintenance of existing accessible loci and induction of accessibility at previously closed regulatory sites constitute essential host epigenomic alterations for infection.
EBV-induced subversion of GC B cell subset regulatory architecture is especially noteworthy. Hyperproliferative cells (c6) mirror GC DZ B cells through elevated levels of LMO2, AICDA, and proliferation genes linked to EBNA-associated accessible chromatin. Likewise, NF-κB activated cells (c2) reflect GC LZ cell chromatin linked to expression of CD83, CD86, and EPHB1. Post-GC effector states (c5) recapitulate in vivo regulatory control of differentiated B cell hallmarks (PRDM1, CD38). Most of these and other GC-associated genes are linked to regions that overlap EBNA footprints. Downregulation of BCL6, a distinctive feature of EBV-induced B cell dynamics relative to GCs in vivo, prevents oncogene repression,104 thereby facilitating latency and priming malignant potential. Although EBV induces several genes that are repressed by BCL6 (e.g., BCL2, MYC), the extent of EBNA binding at GC regulatory loci indicates a previously unappreciated degree of viral epigenomic manipulation of GC dynamics. EBV-induced EPHB1 and CD274 (PD-L1) expression and CXCR4 downregulation may enable checkpoint surveillance bypass by mitigating potentially suppressive interactions with other immune cells involved in GC progression. Along with immunoevasive functions of EBV latency proteins, these phenotypic characteristics would facilitate viral access to and persistence in MBCs.
Transcription factor motif analysis provides further evidence of virus-induced establishment of GC regulatory dynamics. Among these were significant reductions in YY1 and BHLHE40 motif accessibility in the transition to the LZ state and the adoption of MBC-like chromatin architecture. Differential YY1 motif accessibility between non-GC and LZ states is noteworthy given YY1’s foundation role in gene regulation and cellular identity via promoter-enhancer looping.105 Likewise, accessibility loss at BHLHE40 sites upon GC engagement is consistent with the protein’s role in restraining GC entry.71 This multiomic dataset thus provides a powerful resource for future investigations of cell state variation dependent on combinatorial motif accessibility and corresponding TF expression. As a demonstration, we validated the functional significance of TFs with predicted roles in EBV-induced GC regulation from cluster-resolved motif accessibility using reverse genetics. Specific loci predicted to regulate gene expression in cis were similarly validated.
Depending on the phenotype comparison, ~10%–35% of DEGs were DAP-linked. This range was similar to the frequencies of differentially accessible motifs for expressed TFs (23%) and DEGs associated with differentially accessible promoters (25%) identified in dexamethasone-treated A549 cells using sci-CAR.28 These similar frequencies in response to diverse stimuli (e.g., glucocorticoid receptor activation, viral infection) raise intriguing questions regarding the fundamental frequency of cellular response genes regulated by accessibility changes. The observed proportion implies that most DEGs may be regulated by higher order chromatin interactions and differential recruitment of transcriptional activators and/or RNA Pol II.
The crisp-ATAC method provides a simple and flexible approach to map ChIP-seq profiles to phenotypes discovered from scATAC-seq. Thus, it can be used to bootstrap scATAC-seq resolution to other omics datasets. When data from suitable reference samples are available, crisp-ATAC can predict phenotype-resolved regulation by evaluating simple or complex combinations of binding sites and histone modifications across differentially accessible sites. In our case, cell-matched empirical scRNA-seq data can be cross-referenced with crisp-ATAC outputs. We expect this method is generally adaptable for comparing cell phenotypes in contexts such as development and responses to stimuli or drugs. This approach should be particularly powerful for exploring time-resolved TF-associated control of cellular behaviors.
Limitations of the study
This study has several limitations. We did not capture aspects of host-virus dynamics acting at other molecular levels. Examples include DNA methylation, three-dimensional chromatin architecture, changes in protein abundance, post-translational modifications, protein-protein interactions, and signaling activity. Although we present a method for inferring DAP-linked TF binding and epigenetic modifications based on empirical scATAC-seq data, we do not have direct ChIP-seq measurements at the single-cell level.
The crisp-ATAC method also has notable constraints. This approach is limited by the availability of ChIP-seq data from appropriate reference samples. Moreover, regulatory predictions must be empirically tested to validate potential functions in gene expression control. Finally, distance limits imposed for identifying cis-regulatory linkages preclude identification of distal gene regulatory elements formed via long-range three-dimensional (3D) nuclear conformation.
STAR★METHODS
RESOURCE AVAILABILITY
Lead contact
Further information and requests should be directed to and will be fulfilled by the Lead Contact, Micah A. Luftig (micah.luftig@duke.edu).
Materials availability
Cas9-RNP-generated knockout LCLs used in this study are available upon request from the Lead Contact.
Data and code availability
Single-cell ATAC-seq data have been deposited in the NIH Sequence Read Archive (SRA: SRP346796) and Gene Expression Omnibus (GEO: GSE189141). These data are publicly available through the accession numbers and identifiers listed in the key resources table.
Data analysis was performed in R using built-in functions and software packages described in the key resources table. R code used for analysis and visualization is available on Zenodo (https://doi.org/10.5281/zenodo.8125208). Additional information related to data analysis is available upon request.
Additional information and requests for data resources should be directed to and will be fulfilled by the lead contact.
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
Anti CD27-APC | BioLegend (mouse anti-Human CD27, clone M-T271) | Cat. #356410; RRID: AB_2561957 |
Anti CD274-PE | BioLegend (mouse anti-Human CD274, clone 29E-2A3) | Cat. #329706; RRID: AB_940368 |
Anti CD46-APC | BioLegend (mouse anti-Human CD46, clone TRA-2-10) | Cat. #352405; RRID: AB_2564356 |
Anti CD46-PE | BioLegend (mouse anti-Human CD46, clone TRA-2-10) | Cat. #352402; RRID: AB_10895756 |
Anti ICAM1-Pacific Blue | BioLegend (mouse anti-Human ICAM1, clone HCD54) | Cat. #322716; RRID: AB_893384 |
Anti NRF2 | Invitrogen (rabbit anti-Human NRF2, polyclonal) | Cat. #PA5-27882; RRID: AB_2545358 |
Anti IgG | Sigma Aldrich (goat anti-Rabbit IgG, polyclonal) | Cat. #12-348 |
Bacterial and virus strains | ||
Epstein-Barr virus (EBV), B95-8 strain | Luftig Lab (Original publication: Johannsen et al., 2004).31 | https://doi.org/10.1073/pnas.0407320101 |
Biological samples | ||
Whole blood samples | Gulf Coast Regional Blood Center (Houston, TX) | N/A |
Chemicals, peptides, and recombinant proteins | ||
TrueCut™ Cas9 Protein v2 | ThermoFisher Scientific | Cat. #A36499 |
Histopaque®-1077 | Sigma-Aldrich (Raleigh, NC) | Cat. #H8889 |
Critical Commercial Assays | ||
BD iMag B cell Negative Selection Kit | BD Biosciences (Franklin Lakes, NJ) | Cat. #558007 |
10x Chromium Next GEM Single | 10x Genomics (Pleasanton, CA) | Prod. #1000285 |
Cell Multiome ATAC + Gene Expression Kit (2x) | ||
Dual Index Kit TT Set A | 10x Genomics (Pleasanton, CA) | Prod. #1000215 |
Single Index Kit N Set A | 10x Genomics (Pleasanton, CA) | Prod. #1000212 |
10x Chromium Chip J | 10x Genomics (Pleasanton, CA) | Prod. #1000230 |
NovaSeq S2 Reagent Kit v1.5 | Illumina (San Diego, CA) | Cat. #20028314 |
NuPage 4–12% Bis-Tris Gel | ThermoFisher Scientific (Invitrogen) | Cat. #NP0322 |
PageRuler Pre-stained Protein Ladder, 10–250 kDa | ThermoFisher Scientific | Cat. #26619 |
Revert 700 Total Protein Stain | Li-COR (Lincoln, NE) | Cat. #926-11011 |
SuperSignal West Femto | ThermoFisher Scientific | Cat. #34096 |
Gene Knockout Kit v2 (See Table S3) | Synthego (Redwood, CA) | N/A |
Synthetic sgRNA Kit (See Table S3) | Synthego (Redwood, CA) | N/A |
Neon™ Transfection System 10uL Kit | ThermoFisher Scientific | Cat. #MPK1096 |
Deposited data | ||
Donor 1 uninfected B cell scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695143; SRX13167303 |
Donor 2 uninfected B cell scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695151; SRX13167311 |
Donor 1 B cells day 2 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695145; SRX13167305 |
Donor 2 B cells day 2 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695153; SRX13167313 |
Donor 1 B cells day 5 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695147; SRX13167307 |
Donor 2 B cells day 5 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695155; SRX13167315 |
Donor 1 B cells day 8 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695149; SRX13167309 |
Donor 2 B cells day 8 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695157; SRX13167317 |
Donor 1 uninfected B cell scRNA-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695144; SRX13167304 |
Donor 2 uninfected B cell scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695152; SRX13167312 |
Donor 1 B cells day 2 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695146; SRX13167306 |
Donor 2 B cells day 2 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695154; SRX13167314 |
Donor 1 B cells day 5 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695148; SRX13167308 |
Donor 2 B cells day 5 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695156; SRX13167316 |
Donor 1 B cells day 8 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695150; SRX13167310 |
Donor 2 B cells day 8 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695158; SRX13167318 |
GM12878 ATAC-seq | Buenrostro et al., 2013126 (GSE47753) | GSM1155957 |
GM12878 EBNA1 ChIP-seq | Tempera et al., 201679 (GSE73887) | GSM1905009 |
GM12878 EBNALP ChIP-seq | Portal et al., 201380 (GSE49338) | GSM1197603 |
Mutu III EBNA2 ChIP-seq | McLellan et al., 201344 (GSE47629) | GSM1153765 |
Mutu III EBNA3C ChIP-seq | McLellan et al., 201344 (GSE47629) | GSM1153766 |
GM12878 IgG control ChIP-seq | Gunnell et al., 2016127 (GSE76869) | GSM2039169 |
GM12878 RelA ChIP-seq | Zhao et al., 201482; Iannetti et al., 2014 81 (GSE55105) | GSM2628088 |
GM12878 RelB ChIP-seq | Zhao et al., 201482; Iannetti et al., 201481 (GSE55105) | GSM2628090 |
GM12878 cRel ChIP-seq | Zhao et al., 201482; Iannetti et al., 201481 (GSE55105) | GSM2628092 |
GM12878 CTCF ChIP-seq | Lee et al., 2012128 (GSE32883) | GSM489290 |
GM12878 RNA Pol II ChIP-seq | Raha et al., 201085 (GSE19550) | GSM487431 |
GM12878 H2AZ ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733767 |
GM12878 H3K4me1 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733772 |
GM12878 H3K4me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733708 |
GM12878 H3K9ac ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733667 |
GM12878 H3K9me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733664 |
GM12878 H3K27ac ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733771 |
GM12878 H3K27me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733758 |
GM12878 H3K36me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733679 |
GM12878 Hi-C | Rao et al., 2014129 (GSE63525) | GSE63525_GM12878_dilution_combined_30.hic |
Oligonucleotides | ||
GAGAAACAUGUCCAUAUAUA | Synthego sgRNA | sgRNA_CD46_1 |
AACUCGUAAGUCCCAUUUGC | Synthego sgRNA | sgRNA_CD46_2 |
UUGCUCCUUAGAGGAAAUAA | Synthego sgRNA | sgRNA_CD46_3 |
GGCCAAGCACGAGAACACUC | Synthego sgRNA | sgRNA_BHLHE40_1 |
CGAGACCACCCGGUGGAGGU | Synthego sgRNA | sgRNA_BHLHE40_2 |
CAGCUCUCCGGCCAAAGGUU | Synthego sgRNA | sgRNA_BHLHE40_3 |
GGAGGUCGAUGUGUUACACC | Synthego sgRNA | sgRNA_MEF2C_1 |
GAGCCAGUGGCAAUAGGUUG | Synthego sgRNA | sgRNA_MEF2C_2 |
GAGACUGGCAUCUCGAAGUU | Synthego sgRNA | sgRNA_MEF2C_3 |
UAGUUGUAACUGAGCGAAAA | Synthego sgRNA | sgRNA_NFE2L2_1 |
GCGACGGAAAGAGUAUGAGC | Synthego sgRNA | sgRNA_NFE2L2_2 |
AUUUGAUUGACAUACUUUGG | Synthego sgRNA | sgRNA_NFE2L2_3 |
UGGGCAGAUCCCGGACCCUU | Synthego sgRNA | sgRNA_JDP2_1 |
CCGAGCUGGGGAGCCCGGUC | Synthego sgRNA | sgRNA_JDP2_2 |
CAAUCAUGGCCCCGAGGUUG | Synthego sgRNA | sgRNA_JDP2_3 |
AAGUAAGUUGAUCUCACCUA | Synthego sgRNA | sgRNA_CD274_dss_1 |
UCCACCAAUUAGAGCACGGC | Synthego sgRNA | sgRNA_CD274_dss_2 |
ACUCAAUGAACCACGAGAGG | Synthego sgRNA | sgRNA_CD274_dss_3 |
UGGGUCUCUGGAAUCACGAC | Synthego sgRNA | sgRNA_CD274_ctrl_1 |
GCGGUGUUGCCCAUGAGAUA | Synthego sgRNA | sgRNA_CD274_ctrl_2 |
TTCAGTGTAATTCTTCTCTGAGCA | IDT | BHLHE40 Forward Primer |
AGGAACGGGGAGCTGATCA | IDT | BHLHE40 Reverse Primer |
AGGGAGGACGAGCAGAAGTA | IDT | MEF2C Forward Primer |
TGGAGTCTAATGAAATGGGGCT | IDT | MEF2C Reverse Primer |
CAGGAGGCTGAGGTTGGAAA | IDT | NFE2L2 Forward Primer |
AACCATTTGTGACTTTGCCCT | IDT | NFE2L2 Reverse Primer |
TGCCTATGATTGGGTCAAGCA | IDT | JDP2 Forward Primer |
GCCCAGTGCTTCTCAAGTGT | IDT | JDP2 Reverse Primer |
Software and algorithms | ||
CellRanger (cellranger-arc) | 10x Genomics (Pleasanton, CA); functions used: cellranger-arc mkref, cellranger-arc mkfastq, cellranger-arc count | v2.0 |
R/R Studio | Open Source/RStudio, PBC (Boston, MA) | v4.0/v1.4.1106 |
Seurat (R package) | Macosko et al., 2015108; Satija et al., 2015109; Stuart et al., 2019110 | v4.0 |
Signac (R package) | Stuart et al.,202134 | v1.4.0 |
Monocle3 (R package) | Qiu et al., 2017130 | v0.2.3 |
Bioconductor (R package) | bioconductor.org; R libraries used include: GenomeInfoDB, ensembldb, EnsDb.Hsapiens.v86, BSgenome.Hsapiens.UCSC.hg38, motifmatcher, JASPAR2020, TFBSTools) | v3.14 |
macs2/macs3 | Liu, 2014113 | v2.2.7/v3.0 |
bedtools | Quinlan and Hall, 2010117; functions used: multiinter | |
GREAT | McLean et al., 201033; http://great.stanford.edu/public/html/ | v4.0.4 |
hicExplorer | Ramirez et al., 2018118; Wolff et al., 2018119; Wolff et al., 2020120 | v3.7.2 |
IGV | Robinson et al., 2011121 | v2.9.2 |
Juicebox | Durand et al.,2016122 | v1.5.1 |
Cytoscape | Shannon et al., 2003123 | v3.8.2 |
Other | ||
RPMI 1640 | Sigma-Aldrich (Raleigh, NC) | Cat. #11875093 |
FBS, heat inactivated | Corning (Durham, NC) | Cat. #35-010-CV |
DMSO | Sigma-Aldrich (Raleigh, NC) | Cat. #D2660-100ML |
10x Chromium Controller | 10x Genomics (Pleasanton, CA) | Prod. #1000204 |
NovaSeq 6000 | Illumina (San Diego, CA) | Cat. #20012850 |
BD FACS Canto II | BD Biosciences (Franklin Lakes, NJ) | N/A |
Sony SH800 Sorter | Sony Biotechnology | N/A |
Neon™ Transfection System | ThermoFisher Scientific | Cat. #MPK5000 |
Human (hg38) reference genome | Genome Reference Consortium | GRCh38.p13 |
Type 1 EBV reference genome | de Jesus et al. 2003131; FASTA accessed via NCBI | NC_007605.1 |
hg38+EBV reference genome | Generated via concatenation of the genomes listed above | N/A |
EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
Ethics statement
The experiments herein were conducted with approval from a Duke University institutional review board (eIRB# Pro00006262). All blood was obtained from de-identified donors via Gulf Coast Regional Blood Center (Houston, TX) and thus no data on donor age or sex was obtained for this study. No protected health information (PHI) or HIPAA identifiers were provided with blood samples. Thus, the experiments reported here were classified as non-human subjects research.
Primary cells
PBMCs and B cells were isolated from de-identified human donor whole blood as described in the method details section.
METHOD DETAILS
B cells and primary EBV infection
B cells were isolated from peripheral blood mononuclear cells (PBMCs) prepared from whole blood (50 mL per donor) as previously described.30 Briefly, total B cells (naive and memory) were enriched via negative isolation. Uninfected samples (Day 0) were cryopreserved in freezing media (10% DMSO +90% FBS), and primary infections with EBV were performed on remaining donor cells at a multiplicity of infection (MOI) of 5 to ensure viral genome delivery to virtually all cells.9 Samples from each infection timepoint were the cryopreserved as done for uninfected samples. Prior to sequencing, cells were analyzed for B cell enrichment via flow cytometry using anti-CD19 staining (>90% for all timepoints) and successful EBV infection via anti-CD23 staining as previously described.30
Preparation of scATAC libraries
Cryopreserved samples from each early infection timepoint of interest were simultaneously thawed by donor and purified to >90% viable cells by Ficoll gradient separation. Viable cells from each timepoint and donor were then prepared as single-cell matched gene expression (scRNA) and chromatin accessibility (scATAC) libraries by the Duke Molecular Genomics Core staff with the 10x Chromium Next GEM Single Cell Multiome ATAC + Gene Expression Kit (10x Genomics, Pleasanton, CA).106,107 Briefly, nuclei were isolated from each sample and subjected to transposition at accessible chromatin sites. Next, transposed nuclei, barcoding master mix, and gel beads containing unique barcode sequences were prepared into single-cell GEMs (Gel bead emulsions) using the Chromium Controller and Chip J. Within each GEM, poly-adenylated (poly-A) mRNA transcripts from individual nuclei were captured by barcoded, indexed poly-T primers and reverse transcribed into cDNA. Simultaneously, a separate barcoded sequence containing a spacer and Illumina P5 adaptor sequence was added to transposed regions within the captured nucleus. Barcoded multiomes were then purified, pooled, and pre-amplified by PCR prior to library construction. The scATAC-seq library for each sample is generated by PCR amplification and incorporation of sample index and Illumina P7 adaptor sequences. The scRNA library construction, sequencing, analysis, and results (have been described previously.30
Sequencing, alignment, and count matrix generation
8 paired-end scATAC-seq libraries (4 timepoints per 2 donors) were pooled onto two lanes of an Illumina S2 flow cell and sequenced at a target depth of 25,000 reads per cell on an Illumina NovaSeq (Illumina, San Diego, CA). Sequencing runs were performed by staff at the Duke Center for Genomic and Computational Biology. Raw base calls for each assay were prepared as sample-demultiplexed FASTQ files using cellranger-arc mkfastq (Cellranger, 10x Genomics), a wrapper of the Illumina bcl2fastq function. Next, sample-matched reads were aligned against genomic references to produce multiome count matrices using cellranger-arc count. One set of count matrices was generated by mapping reads to a concatenated genomic reference constructed from the human genome (GRCh38) with the ~172 kB type 1 EBV genome (NC_007605) included as an extra chromosome. These outputs, as well as a second set of count matrices generated by mapping to GRCh38 only were used for downstream joint RNA and ATAC analyses. Compatible reference packages were assembled from the relevant genome (.fa) and annotation (.gtf) files using cellranger-arc mkref.
Data QC and scMultiome analysis
Direct analysis of scRNA- and scATAC-seq data was conducted in R using Signac,34 an extension of Seurat.108-110 Following read mapping and counting, scATAC-seq data (linked to scRNA measurements) were obtained from between 8,934 and 20,000 cells per sample. After QC filtering, a total of 52,271 and 44,920 cells were analyzed across the infection timecourse for donors TX1241 and TX1242, respectively. Peaks across all samples were normalized using latent semantic indexing (LSI) as previously described.111 This process consists of Term Frequency-Inverse Document Frequency (TF-IDF) normalization to reduce bias from sequencing depth variation and enhance rare peak detection followed by singular value decomposition (SVD) of the normalized matrix.34 TF motif enrichment matrices generated as direct outputs from cellranger-arc count were also analyzed. Pseudotime graphs were calculated for scATAC and motif accessibility by preparing the assays as cell datasets, performing dimensional reduction, and using the learn_graph function in Cicero (for peaks) and Monocle3 (for motifs).32,112 For multimodal analysis, host chromatin accessibility and gene expression were analyzed for each separate timepoint and for a merged object containing Day 0 and Day 8 multiome data. Nucleosome signal and transcription start site (TSS) enrichment were calculated and used for QC filtering (Nucleosome.signal <2, TSS.enrichment >1). ATAC peaks were called using macs2113 with hg38 annotations. Gene expression data in the joint analysis was processed as described for the RNA-only analysis apart from using Signac’s SCTransform function instead of log normalization for expression counts. Top differential features in each assay (‘peaks’ and ‘SCT’) were determined, and multimodal clustering via weighted nearest neighbors and UMAP dimensional reduction were performed for integrated data visualization. Cluster identities defined in the RNA-only assay were mapped to the merged joint dataset, which contained cells representing all identified subpopulations. Peaks with significant (anti−) correlation (p < 0.05 for Z score of correlation coefficients) to differentially expressed genes were identified using the LinkPeaks function in Signac, which was informed by SHARE-seq.114 Biological zero-preserving read imputation of scRNA data was also performed using a Seurat wrapper for adaptive low-rank approximation (RunALRA) to correct for technical dropout.115 Gene ontology (GO) biological process enrichment for DAP-linked DEGs from phenotype-specific comparisons were generated using Seurat FindMarkers outputs in conjunction with gene list analysis (enrichGO, dotplot) using the clusterProfiler software package.116
crispATAC workflow and reference data curation
ChIP-referenced inference from single-cell phenotype ATAC (crispATAC) was developed to predict subpopulation-resolved gene regulatory features. In a typical workflow, cluster-level chromatin accessibility tracks are cross-referenced against ChIP-Seq (Chromatin Immunoprecipitation Sequencing) profiles for epigenetic marks and TFs of interest measured from a reference cell phenotype (in this study, lymphoblastoid cell lines such as GM12878) via multiple intersection. In this study, cluster-specific called peaks from the multimodal dataset were extracted and prepared as simplified genomic range files (3-column.bed file format). Next, the desired ChIP-Seq datasets for the reference phenotype were downloaded and, where applicable, converted to.bedgraph format to be used as input for peak calling with the macs2 function bdgpeakcall with default parameters. The ChIP-seq (and Hi-C) datasets used for crispATAC in this study are all publicly available from the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO). Once all ATAC-seq and ChIP-seq peak files were generated, all were used as inputs to a single call of the bedtools117 function multiinter with default parameters, which output a matrix of all genomic range intersection intervals where at least one input file exhibited a peak. This intersection matrix was imported to R as a data frame and analyzed to identify common and/or differential intervals (matrix rows) among scATAC-seq cluster phenotypes, epigenetic marks, and TFs using Boolean logic gating by dataset (matrix columns). For a given crispATAC recipe (e.g., peaks in cluster 1 not in cluster 2 intersected with EBNA2 ChIP peaks = [c1 ! c2] ∩ EBNA2), the genome intervals matching the gating criteria were returned and converted to.bed files. Lists of differentially accessible TF-associated sites generated in this way were subsequently analyzed with the Genomic Regions Enrichment of Annotations Tool (GREAT) 33 to identify single-nearest cis-regulated genes within 1 megabase of each query site. As a final step, output lists of potential linked genes were intersected with the top marker genes identified from the corresponding cluster-wise comparison in the scRNA-seq assay using the Seurat FindMarkers function. This process yielded integration of direct scRNA-seq and ATAC-seq measurements with subpopulation-resolved regulatory inferences from ensemble ChIP profiles. In a similar but separate approach, scATAC and ChIP peaks were intersected with topologically associated domain (TAD) boundaries (prepared using hicExplorer 118-120) and nuclear subcompartments from GM12878 Hi-C data to study differentially accessible TF-associated sites in the context of 3D nuclear architecture.
Visualization of crispATAC outputs, gene ontologies, and networks
Data for genes of interest identified from crispATAC recipes were explored using dimensionally reduced (UMAP) expression maps and cluster-level accessibility tracks (CoveragePlot function in Signac34), called peaks aligned with TFs and epigenetic marks (IGV,121), and local neighborhoods in Hi-C contact maps (Juicebox122). Cluster-resolved gene ontologies were generated and quantified by GREAT.33 Top scRNA assay cluster markers and GREAT output gene lists were also visualized as annotated networks using Cytoscape.123
Cas9/RNP-based functional analysis and flow cytometry
To evaluate the functional role of transcription factors whose motif accessibilities differed between the GC LZ NF-κB activated (JDP2 and NFE2L2) and non-GC, more differentiated (BHLHE40 and MEF2C) state, we generated knockouts by transfecting Cas9-ribonucleoprotein (RNP) complexes targeting these genes of interest. Briefly, to assemble the complexes, TrueCut Cas9 protein-v2 (ThermoFisher, cat. #A36498) was incubated with both sgRNA (Gene Knockout Kit v2, Synthego) targeting the gene of interest and CD46, in protein/sgRNA molar ratio of 1:3. Cells to be transfected were resuspended in Buffer R (Neon System Transfection Kit). The transfection was performed using the ThermoFisher Neon Transfection System (10 μL kit) with the following setup: 1 pulse, 1350 mV for 30 ms. Loss of expression of CD46,76 measured by FACS, was used as a proxy to identify KO cells. Additionally, KO LCLs were validated by Sanger sequencing. Primers flanking the regions targeted by Cas9 sgRNAs used to generate BHLHE40, JDP2, MEF2C, and NFE2L2 KO LCL (Integrated DNA Technologies, IDT) were used for PCR amplification of sgRNA target regions. Amplified PCR products were sent for sequencing and analysis (Synthego) to confirm successful lesion induction based on the type and predicted frequency of indels.124
LCLs were stained using α-CD46-PE (BioLegend cat. #352402), α-ICAM1-Pacific Blue (BioLegend cat. #322716), and α-CD27-APC (BioLegend cat. #356410) antibodies. CD46−/ICAM1Hi/CD27Lo cells were sorted using a Sony SH800 Cell Sorter. Post-sort checking and tracking of CD46−/ICAM1Lo/CD27Hi populations was performed on a BD FACS Canto II analyzer system.
To analyze the role of the putative enhancer located between PDCDL1G2 and CD274, we used the same Cas9-RNP approach described above, to generate the deletion of this site. More specifically, multiple sgRNAs (gCD274_dss) targeting this enhancer were used and as a control, a 7kb-apart from the queried peak was targeted with sgRNAs (gCD724_ctrl). CD274 expression was analyzed in CD46 negative cells. The cells were stained using α-CD46-APC (BioLegend cat. #352405) and α-CD274-PE (BioLegend cat. #329706).
Western blot analysis
Based on reduced predicted indel frequency for NFE2L2 (NRF2) replicate LCLs, protein expression levels of NRF2 were evaluated by Western blot. Cell lysates were prepared from WT and NFE2L2 KO LCL replicates with and without hydrogen peroxide treatment (400 mM H2O2 for 3 h @ 37°C) to stabilize NRF2 protein, which is normally constitutively targeted for degradation in the absence of oxidative stress.125 Lysates were sonicated, total protein was quantified by Bradford assay, and a total of 30 μg protein per sample with added 2-mercaptoethanol and loading buffer were run on 4–12% Bis-Tris gels (Invitrogen NuPage 10-well 1.0–1.5mm gel, ThermoFisher cat. #NP0322BOX). Electrophoresis was performed using 1x MES buffer at a running voltage of 120 V for 90 min, using 10–250 kDa PageRuler protein ladder (Thermo Scientific, cat. #26619) as a size standard. Electrophoresed proteins were transferred to PVDF membranes using a TransBlot Turbo Transfer System at 25 V, 1.3 A, for 12 min (Bio-Rad). After transfer, blots were imaged using a Li-COR Odyssey imager (Li-COR Biosciences, Lincoln, NE) after incubation with total protein stain (Li-COR Revert 700 Total Protein Stain) and washing (1x TBST, 3 washes for 5 min each). After revert imaging, blots were blocked with 5% milk in 1x TBST for 1 h at room temperature, washed with TBST, and incubated overnight at 4°C with 1:2000 primary antibody (rabbit anti-human NRF2, Invitrogen, Thermo Fisher cat. #PA5-27882) in 5% BSA with sodium azide. Primary antibody-incubated blots were washed, incubated with 1:3500 goat anti-rabbit IgG HRP-conjugated secondary antibody (Sigma Aldrich, prod. #12–348) in 1x TBST with 5% milk for 1 h at room temperature, washed, incubated with HRP substrate (Thermo Scientific SuperSignal West Femto, cat. #34096), and imaged for NRF2 and protein ladder detection using a Li-COR Odyssey imager (600 and chemiluminescent channels).
QUANTIFICATION AND STATISTICAL ANALYSIS
Statistical analysis
Differentially accessible peaks (DAPs) between scATAC-seq cell clusters were identified using the FindMarkers() function in Seurat/Signac, which calculates raw and Bonferroni-adjusted p values from logistic regression differential accessibility testing of peak intensities. Outputs from this analysis for comparisons of interest are provided in supplementary files. DAPs significantly correlated (p < 0.05 for Z score of correlation coefficients) with differentially expressed genes (DEGs) were identified using the LinkPeaks() function in Signac, which was informed by SHARE-seq.114 Significance of differential gene expression between cell clusters was evaluated by Bonferroni-corrected Wilcoxon rank-sum tests and Kolmogorov-Smirnov (KS) tests. Gene ontology (GO) biological process enrichment for DAP-linked DEGs from phenotype-specific comparisons were generated using Seurat FindMarkers() outputs in conjunction with gene list analysis (enrichGO, dotplot) using the clusterProfiler software package.116 For FACS studies of knockout LCLs, statistically significant differences in gated population frequencies and mean fluorescence intensity (MFI) were evaluated via Two- and One-way ANOVA followed by post-hoc Dunnett method, respectively.
Supplementary Material
Highlights.
scATAC-seq of B cell and viral chromatin accessibility in primary EBV infection
Growth-arrested EBV+ cells develop heterochromatin at key B cell activation loci
Successful infection actuates GC-like B cell chromatin dynamics without BCL6
Gene and regulatory locus multiomic predictions validated in knockout studies
ACKNOWLEDGMENTS
We thank the Duke University Molecular Genomics Core (MGC) and Center for Genomic and Computational Biology (GCB), especially Emily Hocke, Karen Abramson, Dr. Simon Gregory, and Dr. Nicolas Devos. We also thank the anonymous donors whose blood donations made this work possible. E.D.S. acknowledges support from the Department of Molecular Genetics and Microbiology Viral Oncology Training Grant (NIH T32 #T32CA009111) and the American Cancer Society - Charlotte County, Virginia TPAC Postdoctoral Fellowship (PF-21-084-01-DMC). This work was supported by the National Institute of Dental and Craniofacial Research (NIDCR; award #R01DE025994).
Footnotes
DECLARATION OF INTERESTS
The authors declare no competing interests.
SUPPLEMENTAL INFORMATION
Supplemental information can be found online at https://doi.org/10.1016/j.celrep.2023.112958.
REFERENCES
- 1.Masucci MG, and Ernberg I (1994). Epstein-Barr virus: adaptation to a life within the immune system. Trends Microbiol. 2, 125–130. [DOI] [PubMed] [Google Scholar]
- 2.Rickinson A, and Kieff E (2007). Epstein-Barr virus, pp. 2655–2700. Fields virology 2. [Google Scholar]
- 3.Raab-Traub N. (2007). EBV-induced oncogenesis. Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis. [Google Scholar]
- 4.Gross AJ, Hochberg D, Rand WM, and Thorley-Lawson DA (2005). EBV and systemic lupus erythematosus: a new perspective. J. Immunol 774, 6599–6607. [DOI] [PubMed] [Google Scholar]
- 5.Bjornevik K, Cortese M, Healy BC, Kuhle J, Mina MJ, Leng Y, Elledge SJ, Niebuhr DW, Scher AI, Munger KL, and Ascherio A (2022). Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 375, 296–301. [DOI] [PubMed] [Google Scholar]
- 6.Lanz TV, Brewer RC, Ho PP, Moon J-S, Jude KM, Fernandez D, Fernandes RA, Gomez AM, Nadj G-S, Bartley CM, et al. (2022). Clonally Expanded B Cells in Multiple Sclerosis Bind EBV EBNA1 and GlialCAM. Nature 603, 321–327. 10.1038/s41586-022-04432-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Roughan JE, and Thorley-Lawson DA (2009). The intersection of Epstein-Barr virus with the germinal center. J. Virol 83, 3968–3976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Dai J, Heckenberg E, Song L, Crawford GE, and Luftig MA (2020). Epstein-Barr Virus Promotes Survival through Germinal Center Light Zone Chromatin Architecture. Preprint. bioRxiv [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Nikitin PA, Yan CM, Forte E, Bocedi A, Tourigny JP, White RE, Allday MJ, Patel A, Dave SS, Kim W, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.McFadden K, Hafez AY, Kishton R, Messinger JE, Nikitin PA, Rathmell JC, and Luftig MA (2016). Metabolic stress is a barrier to Epstein–Barr virus-mediated B-cell immortalization. Proc. Natl. Acad. Sci. USA 113, E782–E790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pope JH, Horne MK, and Scott W (1968). Transformation of foetal human leukocytes in vitro by filtrates of a human leukaemic cell line containing herpes-like virus. Int. J. Cancer 3, 857–866. [DOI] [PubMed] [Google Scholar]
- 12.Bird AG, McLachlan SM, and Britton S (1981). Cyclosporin A promotes spontaneous outgrowth in vitro of Epstein-Barr virus-induced B-cell lines. Nature 289, 300–301. [DOI] [PubMed] [Google Scholar]
- 13.SoRelle ED, Dai J, Bonglack EN, Heckenberg EM, Zhou JY, Giamberardino SN, Bailey JA, Gregory SG, Chan C, and Luftig MA (2021). Single-cell RNA-seq reveals transcriptomic heterogeneity mediated by host–pathogen dynamics in lymphoblastoid cell lines. Elife 10, e62586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mrozek-Gorska P, Buschle A, Pich D, Schwarzmayr T, Fechtner R, Scialdone A, and Hammerschmidt W (2019). Epstein-Barr virus reprograms human B lymphocytes immediately in the prelatent phase of infection. Proc. Natl. Acad. Sci. USA 116, 16046–16055. 10.1073/pnas.1901314116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wang C, Li D, Zhang L, Jiang S, Liang J, Narita Y, Hou I, Zhong Q, Zheng Z, Xiao H, et al. (2019). RNA sequencing analyses of gene expression during Epstein-Barr virus infection of primary B lymphocytes. J. Virol 93, e00226–19–e00219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lamontagne RJ, Soldan SS, Su C, Wiedmer A, Won KJ, Lu F, Goldman AR, Wickramasinghe J, Tang H-Y, Speicher DW, et al. (2021). A multi-omics approach to Epstein-Barr virus immortalization of B-cells reveals EBNA1 chromatin pioneering activities targeting nucleotide metabolism. PLoS Pathog. 17, e1009208. 10.1371/journal.ppat.1009208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hernando H, Islam ABMMK, Rodríguez-Ubreva J, Forné I, Ciudad L, Imhof A, Shannon-Lowe C, and Ballestar E (2014). Epstein–Barr virus-mediated transformation of B cells induces global chromatin changes independent to the acquisition of proliferation. Nucleic Acids Res. 42, 249–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhou H, Schmidt SCS, Jiang S, Willox B, Bernhardt K, Liang J, Johannsen EC, Kharchenko P, Gewurz BE, Kieff E, and Zhao B (2015). Epstein-Barr virus oncoprotein super-enhancers control B cell growth. Cell Host Microbe 17, 205–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hong T, Parameswaran S, Donmez OA, Miller D, Forney C, Lape M, Saint Just Ribeiro M, Liang J, Edsall LE, Magnusen AF, et al. (2021). Epstein–Barr virus nuclear antigen 2 extensively rewires the human chromatin landscape at autoimmune risk loci. Genome Res. 31, 2185–2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shapiro E, Biezuner T, and Linnarsson S (2013). Single-cell sequencing-based technologies will revolutionize whole-organism science. Nat. Rev. Genet 14, 618–630. [DOI] [PubMed] [Google Scholar]
- 21.Junker JP, and van Oudenaarden A (2014). Every cell is special: genome-wide studies add a new dimension to single-cell biology. Cell 157, 8–11. [DOI] [PubMed] [Google Scholar]
- 22.Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP, Chang HY, and Greenleaf WJ (2015). Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Shalek AK, Satija R, Adiconis X, Gertner RS, Gaublomme JT, Raychowdhury R, Schwartz S, Yosef N, Malboeuf C, Lu D, et al. (2013). Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498, 236–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wills QF, Livak KJ, Tipping AJ, Enver T, Goldson AJ, Sexton DW, and Holmes C (2013). Single-cell gene expression analysis reveals genetic associations masked in whole-tissue experiments. Nat. Biotechnol 31, 748–752. [DOI] [PubMed] [Google Scholar]
- 25.Efremova M, and Teichmann SA (2020). Computational methods for single-cell omics across modalities. Nat. Methods 17, 14–17. [DOI] [PubMed] [Google Scholar]
- 26.Chen S, Lake BB, and Zhang K (2019). High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell. Nat. Biotechnol 37, 1452–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhu C, Yu M, Huang H, Juric I, Abnousi A, Hu R, Lucero J, Behrens MM, Hu M, and Ren B (2019). An ultra high-throughput method for single-cell joint analysis of open chromatin and transcriptome. Nat. Struct. Mol. Biol 26, 1063–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cao J, Cusanovich DA, Ramani V, Aghamirzaie D, Pliner HA, Hill AJ, Daza RM, McFaline-Figueroa JL, Packer JS, Christiansen L, et al. (2018). Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361, 1380–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Clark SJ, Argelaguet R, Kapourani C-A, Stubbs TM, Lee HJ, Alda-Catalinas C, Krueger F, Sanguinetti G, Kelsey G, Marioni JC, et al. (2018). scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun 9, 781–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.SoRelle ED, Dai J, Reinoso-Vizcaino NM, Barry AP, Chan C, and Luftig MA (2022). Time-resolved transcriptomes reveal diverse B cell fate trajectories in the early response to Epstein-Barr virus infection. Cell Rep. 40, 111286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Johannsen E, Luftig M, Chase MR, Weicksel S, Cahir-McFarland E, Illanes D, Sarracino D, and Kieff E (2004). Proteins of purified Epstein-Barr virus. Proc. Natl. Acad. Sci. USA 101, 16286–16291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Pliner HA, Packer JS, McFaline-Figueroa JL, Cusanovich DA, Daza RM, Aghamirzaie D, Srivatsan S, Qiu X, Jackson D, Minkina A, et al. (2018). Cicero predicts cis-regulatory DNA interactions from single-cell chromatin accessibility data. Mol. Cell 71, 858–871.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.McLean CY, Bristor D, Hiller M, Clarke SL, Schaar BT, Lowe CB, Wenger AM, and Bejerano G (2010). GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol 28, 495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Stuart T, Srivastava A, Madad S, Lareau CA, and Satija R (2021). Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ma Y, Walsh MJ, Bernhardt K, Ashbaugh CW, Trudeau SJ, Ashbaugh IY, Jiang S, Jiang C, Zhao B, Root DE, et al. (2017). CRISPR/Cas9 screens reveal Epstein-Barr virus-transformed B cell host dependency factors. Cell Host Microbe 21, 580–591.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Courtois-Cox S, Jones SL, and Cichowski K (2008). Many roads lead to oncogene-induced senescence. Oncogene 27, 2801–2809. [DOI] [PubMed] [Google Scholar]
- 37.Di Micco R, Sulli G, Dobreva M, Liontos M, Botrugno OA, Gargiulo G, Dal Zuffo R, Matti V, d’Ario G, Montani E, et al. (2011). Interplay between oncogene-induced DNA damage response and heterochromatin in senescence and cancer. Nat. Cell Biol 13, 292–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lenain C, de Graaf CA, Pagie L, Visser NL, de Haas M, de Vries SS, Peric-Hupkes D, van Steensel B, and Peeper DS (2017). Massive reshaping of genome–nuclear lamina interactions during oncogene-induced senescence. Genome Res. 27, 1634–1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yanai H, Ban T, Wang Z, Choi MK, Kawamura T, Negishi H, Nakasato M, Lu Y, Hangai S, Koshiba R, et al. (2009). HMGB proteins function as universal sentinels for nucleic-acid-mediated innate immune responses. Nature 462, 99–103. [DOI] [PubMed] [Google Scholar]
- 40.Krynetskaia NF, Phadke MS, Jadhav SH, and Krynetskiy EY (2009). Chromatin-associated proteins HMGB1/2 and PDIA3 trigger cellular response to chemotherapy-induced DNA damage. Mol. Cancer Therapeut. 8, 864–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Stros M, Ozaki T, Bacıková A, Kageyama H, and Nakagawara A (2002). HMGB1 and HMGB2 cell-specifically down-regulate the p53- and p73-dependent sequence-specific transactivation from the human Bax gene promoter. J. Biol. Chem 277, 7157–7164. [DOI] [PubMed] [Google Scholar]
- 42.Glück S, Guey B, Gulen MF, Wolter K, Kang T-W, Schmacke NA, Bridgeman A, Rehwinkel J, Zender L, and Ablasser A (2017). Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat. Cell Biol 19, 1061–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gillman ACT, Parker G, Allday MJ, and Bazot Q (2018). Epstein-Barr virus nuclear antigen 3C inhibits expression of COBLL1 and the ADAM28-ADAMDEC1 locus via interaction with the histone lysine demethylase KDM2B. J. Virol 92, e01362–18–e01318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.McClellan MJ, Wood CD, Ojeniyi O, Cooper TJ, Kanhere A, Arvey A, Webb HM, Palermo RD, Harth-Hertle ML, Kempkes B, et al. (2013). Modulation of enhancer looping and differential gene targeting by Epstein-Barr virus transcription factors directs cellular reprogramming. PLoS Pathog. 9, e1003636. 10.1371/journal.ppat.1003636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kuchen S, Robbins R, Sims GP, Sheng C, Phillips TM, Lipsky PE, and Ettinger R (2007). Essential role of IL-21 in B cell activation, expansion, and plasma cell generation during CD4+ T cell-B cell collaboration. J. Immunol 179, 5886–5896. [DOI] [PubMed] [Google Scholar]
- 46.Bessa J, Kopf M, and Bachmann MF (2010). Cutting edge: IL-21 and TLR signaling regulate germinal center responses in a B cell-intrinsic manner. J. Immunol 184, 4615–4619. [DOI] [PubMed] [Google Scholar]
- 47.Gonzalez DG, Cote CM, Patel JR, Smith CB, Zhang Y, Nickerson KM, Zhang T, Kerfoot SM, and Haberman AM (2018). Nonredundant Roles of IL-21 and IL-4 in the Phased Initiation of Germinal Center B Cells and Subsequent Self-Renewal Transitions. J. Immunol 201, 3569–3579. 10.4049/jimmunol.1500497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Challa-Malladi M, Lieu YK, Califano O, Holmes AB, Bhagat G, Murty VV, Dominguez-Sola D, Pasqualucci L, and Dalla-Favera R (2011). Combined genetic inactivation of β2-Microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma. Cancer Cell 20, 728–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Emilie D, Wallon C, Galanaud P, Fischer A, Olive D, and Delfraissy JF (1988). Role of the LFA3-CD2 interaction in human specific B cell differentiation. J. Immunol 141, 1912–1918. 10.4049/jimmunol.141.6.1912. [DOI] [PubMed] [Google Scholar]
- 50.Hess P, Pihan G, Sawyers CL, Flavell RA, and Davis RJ (2002). Survival signaling mediated by c-Jun NH2-terminal kinase in transformed B lymphoblasts. Nat. Genet 32, 201–205. [DOI] [PubMed] [Google Scholar]
- 51.Francis SH, Blount MA, and Corbin JD (2011). Mammalian cyclic nucleotide phosphodiesterases: molecular mechanisms and physiological functions. Physiol. Rev 91, 651–690. [DOI] [PubMed] [Google Scholar]
- 52.Peter D, Jin SLC, Conti M, Hatzelmann A, and Zitt C (2007). Differential Expression and Function of Phosphodiesterase 4 (PDE4) Subtypes in Human Primary CD4+ T Cells: Predominant Role of PDE4D1. J. Immunol 178, 4820–4831. 10.4049/jimmunol.178.8.4820. [DOI] [PubMed] [Google Scholar]
- 53.Mancao C, and Hammerschmidt W (2007). Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood. The Journal of the American Society of Hematology 110, 3715–3721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Minguet S, Huber M, Rosenkranz L, Schamel WWA, Reth M, and Brummer T (2005). Adenosine and cAMP are potent inhibitors of the NF-κB pathway downstream of immunoreceptors. Eur. J. Immunol 35, 31–41. [DOI] [PubMed] [Google Scholar]
- 55.Green JA, and Cyster JG (2012). S1PR2 links germinal center confinement and growth regulation. Immunol. Rev 247, 36–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Guo R, Zhang Y, Teng M, Jiang C, Schineller M, Zhao B, Doench JG, O’Reilly RJ, Cesarman E, Giulino-Roth L, and Gewurz BE (2020). DNA methylation enzymes and PRC1 restrict B-cell Epstein–Barr virus oncoprotein expression. Nat. Microbiol 5, 1051–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Osthus RC, Karim B, Prescott JE, Smith BD, McDevitt M, Huso DL, and Dang CV (2005). The Myc target gene JPO1/CDCA7 is frequently overexpressed in human tumors and has limited transforming activity in vivo. Cancer Res. 65, 5620–5627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Prescott JE, Osthus RC, Lee LA, Lewis BC, Shim H, Barrett JF, Guo Q, Hawkins AL, Griffin CA, and Dang CV (2001). A novel c-Myc-responsive gene, JPO1, participates in neoplastic transformation. J. Biol. Chem 276, 48276–48284. [DOI] [PubMed] [Google Scholar]
- 59.SoRelle ED, Reinoso-Vizcaino NM, Horn GQ, and Luftig MA (2022). Epstein-Barr virus perpetuates B cell germinal center dynamics and generation of autoimmune-associated phenotypes in vitro. Front. Immunol 13, 1001145. 10.3389/fimmu.2022.1001145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Thorley-Lawson DA, Hawkins JB,Tracy SI, and Shapiro M (2013). The pathogenesis of Epstein–Barr virus persistent infection. Curr. Opin. Virol. 3, 227–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Roughan JE, Torgbor C, and Thorley-Lawson DA (2010). Germinal center B cells latently infected with Epstein-Barr virus proliferate extensively but do not increase in number. J. Virol 84, 1158–1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Victora GD, and Nussenzweig MC (2022). Germinal centers. Annu. Rev. Immunol. 40, 413–442. [DOI] [PubMed] [Google Scholar]
- 63.Pei Y, Banerjee S, Jha HC, Sun Z, and Robertson ES (2017). An essential EBV latent antigen 3C binds Bcl6 for targeted degradation and cell proliferation. PLoS Pathog. 13, e1006500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Martín-Pérez D, Vargiu P, Montes-Moreno S, León EA, Rodríguez-Pinilla SM, Lisio LD, Martínez N, Rodríguez R, Mollejo M, Castellvi J, et al. (2012). Epstein-Barr virus microRNAs repress BCL6 expression in diffuse large B-cell lymphoma. Leukemia 26, 180–183. [DOI] [PubMed] [Google Scholar]
- 65.Boccellato F, Anastasiadou E, Rosato P, Kempkes B, Frati L, Faggioni A, and Trivedi P (2007). EBNA2 interferes with the germinal center phenotype by downregulating BCL6 and TCL1 in non-Hodgkin’s lymphoma cells. J. Virol 81, 2274–2282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Allen CDC, Ansel KM, Low C, Lesley R, Tamamura H, Fujii N, and Cyster JG (2004). Germinal center dark and light zone organization is mediated by CXCR4 and CXCR5. Nat. Immunol 5, 943–952. [DOI] [PubMed] [Google Scholar]
- 67.Nakayama T, Fujisawa R, Izawa D, Hieshima K, Takada K, and Yoshie O (2002). Human B cells immortalized with Epstein-Barr virus upregulate CCR6 and CCR10 and downregulate CXCR4 and CXCR5. J. Virol 76, 3072–3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Natkunam Y, Zhao S, Mason DY, Chen J, Taidi B, Jones M, Hammer AS, Hamilton Dutoit S, Lossos IS, and Levy R (2007). The oncoprotein LMO2 is expressed in normal germinal-center B cells and in human B-cell lymphomas. Blood 109, 1636–1642. 10.1182/blood-2006-08-039024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pasqualucci L, Bhagat G, Jankovic M, Compagno M, Smith P, Muramatsu M, Honjo T, Morse HC III, Nussenzweig MC, and Dalla-Favera R (2008). AID is required for germinal center–derived lymphomagenesis. Nat. Genet 40, 108–112. [DOI] [PubMed] [Google Scholar]
- 70.Muramatsu M, Sankaranand VS, Anant S, Sugai M, Kinoshita K, Davidson NO, and Honjo T (1999). Specific expression of activation-induced cytidine deaminase (AID), a novel member of the RNA-editing deaminase family in germinal center B cells. J. Biol. Chem 274, 18470–18476. [DOI] [PubMed] [Google Scholar]
- 71.Rauschmeier R, Reinhardt A, Gustafsson C, Glaros V, Artemov AV, Dunst J, Taneja R, Adameyko I, Månsson R, Busslinger M, and Kreslavsky T (2022). Bhlhe40 function in activated B and TFH cells restrains the GC reaction and prevents lymphomagenesis. J. Exp. Med 219, e20211406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Dudziak D, Kieser A, Dirmeier U, Nimmerjahn F, Berchtold S, Steinkasserer A, Marschall G, Hammerschmidt W, Laux G, and Bornkamm GW (2003). Latent membrane protein 1 of Epstein-Barr virus induces CD83 by the NF-κB signaling pathway. J. Virol 77, 8290–8298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Laidlaw BJ, Schmidt TH, Green JA, Allen CDC, Okada T, and Cyster JG (2017). The Eph-related tyrosine kinase ligand Ephrin-B1 marks germinal center and memory precursor B cells. J. Exp. Med 214, 639–649. 10.1084/jem.20161461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lu P, Shih C, and Qi H (2017). Ephrin B1-mediated repulsion and signaling control germinal center T cell territoriality and function. Science 356, eaai9264. 10.1126/science.aai9264. [DOI] [PubMed] [Google Scholar]
- 75.Bräuninger A, Schmitz R, Bechtel D, Rennié C, Hansmann M-L, and Küppers R (2006). Molecular biology of Hodgkin’s and Reed/Sternberg cells in Hodgkin’s lymphoma. Int. J. Cancer 118, 1853–1861. 10.1002/ijc.21716. [DOI] [PubMed] [Google Scholar]
- 76.Akidil E, Albanese M, Buschle A, Ruhle A, Pich D, Keppler OT, and Hammerschmidt W (2021). Highly efficient CRISPR-Cas9-mediated gene knockout in primary human B cells for functional genetic studies of Epstein-Barr virus infection. PLoS Pathog. 17, e1009117. 10.1371/journal.ppat.1009117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Molkentin JD, Black BL, Martin JF, and Olson EN (1996). Mutational analysis of the DNA binding, dimerization, and transcriptional activation domains of MEF2C. Mol. Cell Biol 16, 2627–2636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Yetming KD, Lupey-Green LN, Biryukov S, Hughes DJ, Marendy EM, Miranda JL, and Sample JT (2020). The BHLF1 locus of Epstein-Barr virus contributes to viral latency and B-cell immortalization. J. Virol 94, e01215–20–e01220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Tempera I, De Leo A, Kossenkov AV, Cesaroni M, Song H, Dawany N, Showe L, Lu F, Wikramasinghe P, and Lieberman PM (2016). Identification of MEF2B, EBF1, and IL6R as direct gene targets of Epstein-Barr virus (EBV) nuclear antigen 1 critical for EBV-infected B-lymphocyte survival. J. Virol 90, 345–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Portal D, Zhou H, Zhao B, Kharchenko PV, Lowry E, Wong L, Quackenbush J, Holloway D, Jiang S, Lu Y, and Kieff E (2013). Epstein-Barr virus nuclear antigen leader protein localizes to promoters and enhancers with cell transcription factors and EBNA2. Proc. Natl. Acad. Sci. USA 110, 18537–18542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Iannetti A, Ledoux AC, Tudhope SJ, Sellier H, Zhao B, Mowla S, Moore A, Hummerich H, Gewurz BE, Cockell SJ, et al. (2014). Regulation of p53 and Rb links the alternative NF-κB pathway to EZH2 expression and cell senescence. PLoS Genet. 10, e1004642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zhao B, Barrera LA, Ersing I, Willox B, Schmidt SCS, Greenfeld H, Zhou H, Mollo SB, Shi TT, Takasaki K, et al. (2014). The NF-κB genomic landscape in lymphoblastoid B cells. Cell Rep. 8, 1595–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.ENCODE Project Consortium (2012). An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Jiang S, and Mortazavi A (2018). Integrating ChIP-seq with other functional genomics data. Brief. Funct. Genomics 17, 104–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Raha D, Wang Z, Moqtaderi Z,Wu L, Zhong G, Gerstein M, Struhl K , and Snyder M (2010). Close association of RNA polymerase II and many transcription factors with Pol III genes. Proc. Natl. Acad. Sci. USA 107, 3639–3644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Devergne O, Hatzivassiliou E, Izumi KM, Kaye KM, Kleijnen MF, Kieff E, and Mosialos G (1996). Association of TRAF1, TRAF2, and TRAF3 with an Epstein-Barr virus LMP1 domain important for B-lymphocyte transformation: role in NF-kappaB activation. Mol. Cell Biol 16, 7098–7108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sandberg M, Hammerschmidt W, and Sugden B (1997). Characterization of LMP-1’s association with TRAF1, TRAF2, and TRAF3. J. Virol 71, 4649–4656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Fries KL, Miller WE,and Raab-Traub N(1999).The A20 protein interacts with the Epstein–Barr virus latent membrane protein 1 (LMP1) and alters the LMP1/TRAF1/TRADD complex. Virology 264, 159–166. [DOI] [PubMed] [Google Scholar]
- 89.Eliopoulos AG, Waites ER, Blake SMS, Davies C, Murray P, and Young LS (2003). TRAF1 is a critical regulator of JNK signaling by the TRAF-binding domain of the Epstein-Barr virus-encoded latent infection membrane protein 1 but not CD40. J. Virol 77, 1316–1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Greenfeld H, Takasaki K, Walsh MJ, Ersing I, Bernhardt K, Ma Y, Fu B, Ashbaugh CW, Cabo J, Mollo SB, et al. (2015). TRAF1 coordinates polyubiquitin signaling to enhance Epstein-Barr virus LMP1-mediated growth and survival pathway activation. PLoS Pathog. 11, e1004890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gao J, Xia L, Lu M, Zhang B, Chen Y, Xu R, and Wang L (2012). TM7SF1 (GPR137B): a novel lysosome integral membrane protein. Mol. Biol. Rep 39, 8883–8889. [DOI] [PubMed] [Google Scholar]
- 92.Gan L, Seki A, Shen K, Iyer H, Han K, Hayer A, Wollman R, Ge X, Lin JR, Dey G, et al. (2019). The lysosomal GPCR-like protein GPR137B regulates Rag and mTORC1 localization and activity. Nat. Cell Biol 21,614–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Settembre C, Di Malta C, Polito VA, Garcia Arencibia M, Vetrini F, Erdin S, Erdin SU, Huynh T, Medina D, Colella P, et al. (2011). TFEB links autophagy to lysosomal biogenesis. science 332, 1429–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Martina JA, Chen Y, Gucek M, and Puertollano R (2012). MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy 8, 903–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Nagelkerke A, Sieuwerts AM, Bussink J, Sweep FCGJ, Look MP, Foekens JA, Martens JWM, and Span PN (2014). LAMP3 is involved in tamoxifen resistance in breast cancer cells through the modulation of autophagy. Endocr. Relat. Cancer 21, 101–112. [DOI] [PubMed] [Google Scholar]
- 96.Simpson EH (1951). The interpretation of interaction in contingency tables. J. Roy. Stat. Soc B 13, 238–241. [Google Scholar]
- 97.Trapnell C. (2015). Defining cell types and states with single-cell genomics. Genome Res. 25, 1491–1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Deisenroth C, and Zhang Y (2010). Ribosome biogenesis surveillance: probing the ribosomal protein-Mdm2-p53 pathway. Oncogene 29, 4253–4260. [DOI] [PubMed] [Google Scholar]
- 99.Forte E, and Luftig MA (2009). MDM2-dependent inhibition of p53 is required for Epstein-Barr virus B-cell growth transformation and infected-cell survival. J. Virol 83, 2491–2499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gruhne B, Sompallae R, and Masucci MG (2009). Three Epstein–Barr virus latency proteins independently promote genomic instability by inducing DNA damage, inhibiting DNA repair and inactivating cell cycle checkpoints. Oncogene 28, 3997–4008. [DOI] [PubMed] [Google Scholar]
- 101.Han I, Harada S, Weaver D, Xue Y, Lane W, Orstavik S, Skalhegg B, and Kieff E (2001). EBNA-LP associates with cellular proteins including DNA-PK and HA95. J. Virol 75, 2475–2481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Dellaire G, Ching RW, Ahmed K, Jalali F, Tse KCK, Bristow RG, and Bazett-Jones DP (2006). Promyelocytic leukemia nuclear bodies behave as DNA damage sensors whose response to DNA double-strand breaks is regulated by NBS1 and the kinases ATM, Chk2, and ATR. J. Cell Biol 175, 55–66. 10.1083/jcb.200604009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Shanbhag NM, Rafalska-Metcalf IU, Balane-Bolivar C, Janicki SM, and Greenberg RA (2010). ATM-dependent chromatin changes silence transcription in cis to DNA double-strand breaks. Cell 141, 970–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ci W, Polo JM, Cerchietti L, Shaknovich R, Wang L, Yang SN, Ye K, Farinha P, Horsman DE, Gascoyne RD, et al. (2009). The BCL6 transcriptional program features repression of multiple oncogenes in primary B cells and is deregulated in DLBCL. Blood 113, 5536–5548. 10.1182/blood-2008-12-193037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Weintraub AS, Li CH, Zamudio AV, Sigova AA, Hannett NM, Day DS, Abraham BJ, Cohen MA, Nabet B, Buckley DL, et al. (2017). YY1 is a structural regulator of enhancer-promoter loops. Cell 171, 1573–1588.e28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zheng GXY, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB, Wheeler TD, McDermott GP, Zhu J, et al. (2017). Massively parallel digital transcriptional profiling of single cells. Nat. Commun 8, 14049–14112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Satpathy AT, Granja JM, Yost KE, Qi Y, Meschi F, McDermott GP, Olsen BN, Mumbach MR, Pierce SE, Corces MR, et al. (2019). Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat. Biotechnol 37, 925–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, et al. (2015). Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Satija R, Farrell JA, Gennert D, Schier AF, and Regev A (2015). Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol 33, 495–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM III, Hao Y, Stoeckius M, Smibert P, and Satija R (2019). Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Cusanovich DA, Daza R, Adey A, Pliner HA, Christiansen L, Gunderson KL, Steemers FJ,Trapnell C, and Shendure J (2015). Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914. 10.1126/science.aab1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, and Rinn JL (2014). The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol 32, 381–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Liu T. (2014). Use model-based analysis of ChIP-Seq (MACS) to analyze short reads generated by sequencing protein-DNA interactions in embryonic stem cells. In Stem Cell Transcriptional Networks (Springer), pp. 81–95. [DOI] [PubMed] [Google Scholar]
- 114.Ma S, Zhang B, LaFave LM, Earl AS, Chiang Z, Hu Y, Ding J, Brack A, Kartha VK, Tay T, et al. (2020). Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116.e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Linderman GC, Zhao J, Roulis M, Bielecki P, Flavell RA, Nadler B, and Kluger Y (2022). Zero-preserving imputation of single-cell RNA-seq data. Nat. Commun 13, 192–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Yu G, Wang L-G, Han Y, and He Q-Y (2012). clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS A J. Integr. Biol 16, 284–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Quinlan AR, and Hall IM (2010). BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ramírez F, Bhardwaj V, Arrigoni L, Lam KC, Grüning BA, Villaveces J, Habermann B, Akhtar A, and Manke T (2018). High-resolution TADs reveal DNA sequences underlying genome organization in flies. Nat. Commun 9, 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wolff J, Bhardwaj V, Nothjunge S, Richard G, Renschler G, Gilsbach R, Manke T, Backofen R, Ramírez F, and Grüning BA (2018). Galaxy HiCExplorer: a web server for reproducible Hi-C data analysis, quality control and visualization. Nucleic Acids Res. 46, W11–W16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wolff J, Rabbani L, Gilsbach R, Richard G, Manke T, Backofen R, and Grüning BA (2020). Galaxy HiCExplorer 3: a web server for reproducible Hi-C, capture Hi-C and single-cell Hi-C data analysis, quality control and visualization. Nucleic Acids Res. 48, W177–W184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, and Mesirov JP (2011). Integrative genomics viewer. Nat. Biotechnol 29, 24–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Durand NC, Robinson JT, Shamim MS, Machol I, Mesirov JP, Lander ES, and Aiden EL (2016). Juicebox provides a visualization system for Hi-C contact maps with unlimited zoom. Cell Syst. 3, 99–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, and Ideker T (2003). Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Conant D, Hsiau T, Rossi N, Oki J, Maures T, Waite K, Yang J, Joshi S, Kelso R, Holden K, et al. (2022). Inference of CRISPR edits from Sanger trace data. The CRISPR Journal 5, 123–130. [DOI] [PubMed] [Google Scholar]
- 125.Suzuki T, Muramatsu A, Saito R, Iso T, Shibata T, Kuwata K, Kawaguchi S. i., Iwawaki T, Adachi S, Suda H, et al. (2019). Molecular mechanism of cellular oxidative stress sensing by Keap1. Cell Rep. 28, 746–758.e4. [DOI] [PubMed] [Google Scholar]
- 126.Buenrostro JD, Giresi PG, Zaba LC, Chang HY, and Greenleaf WJ (2013). Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Gunnell A, Webb HM, Wood CD, McClellan MJ, Wichaidit B, Kempkes B, Jenner RG, Osborne C, Farrell PJ, and West MJ (2016). RUNX super-enhancer control through the Notch pathway by Epstein-Barr virus transcription factors regulates B cell growth. Nucleic Acids Res. 44, 4636–4650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lee B-K, Bhinge AA, Battenhouse A, McDaniell RM, Liu Z, Song L, Ni Y, Birney E, Lieb JD, Furey TS, et al. (2012). Cell-type specific and combinatorial usage of diverse transcription factors revealed by genome-wide binding studies in multiple human cells. Genome Res. 22, 9–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Rao SSP, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, Sanborn AL, Machol I, Omer AD, Lander ES, and Aiden EL (2014). A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Qiu X, Mao Q, Tang Y, Wang L, Chawla R, Pliner HA, and Trapnell C (2017). Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.de Jesus O, Smith PR, Spender LC, Elgueta Karstegl C, Niller HH, Huang D, and Farrell PJ (2003). Updated Epstein–Barr virus (EBV) DNA sequence and analysis of a promoter for the BART (CST, BARF0) RNAs of EBV. J. Gen. Virol 84, 1443–1450. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Single-cell ATAC-seq data have been deposited in the NIH Sequence Read Archive (SRA: SRP346796) and Gene Expression Omnibus (GEO: GSE189141). These data are publicly available through the accession numbers and identifiers listed in the key resources table.
Data analysis was performed in R using built-in functions and software packages described in the key resources table. R code used for analysis and visualization is available on Zenodo (https://doi.org/10.5281/zenodo.8125208). Additional information related to data analysis is available upon request.
Additional information and requests for data resources should be directed to and will be fulfilled by the lead contact.
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
Anti CD27-APC | BioLegend (mouse anti-Human CD27, clone M-T271) | Cat. #356410; RRID: AB_2561957 |
Anti CD274-PE | BioLegend (mouse anti-Human CD274, clone 29E-2A3) | Cat. #329706; RRID: AB_940368 |
Anti CD46-APC | BioLegend (mouse anti-Human CD46, clone TRA-2-10) | Cat. #352405; RRID: AB_2564356 |
Anti CD46-PE | BioLegend (mouse anti-Human CD46, clone TRA-2-10) | Cat. #352402; RRID: AB_10895756 |
Anti ICAM1-Pacific Blue | BioLegend (mouse anti-Human ICAM1, clone HCD54) | Cat. #322716; RRID: AB_893384 |
Anti NRF2 | Invitrogen (rabbit anti-Human NRF2, polyclonal) | Cat. #PA5-27882; RRID: AB_2545358 |
Anti IgG | Sigma Aldrich (goat anti-Rabbit IgG, polyclonal) | Cat. #12-348 |
Bacterial and virus strains | ||
Epstein-Barr virus (EBV), B95-8 strain | Luftig Lab (Original publication: Johannsen et al., 2004).31 | https://doi.org/10.1073/pnas.0407320101 |
Biological samples | ||
Whole blood samples | Gulf Coast Regional Blood Center (Houston, TX) | N/A |
Chemicals, peptides, and recombinant proteins | ||
TrueCut™ Cas9 Protein v2 | ThermoFisher Scientific | Cat. #A36499 |
Histopaque®-1077 | Sigma-Aldrich (Raleigh, NC) | Cat. #H8889 |
Critical Commercial Assays | ||
BD iMag B cell Negative Selection Kit | BD Biosciences (Franklin Lakes, NJ) | Cat. #558007 |
10x Chromium Next GEM Single | 10x Genomics (Pleasanton, CA) | Prod. #1000285 |
Cell Multiome ATAC + Gene Expression Kit (2x) | ||
Dual Index Kit TT Set A | 10x Genomics (Pleasanton, CA) | Prod. #1000215 |
Single Index Kit N Set A | 10x Genomics (Pleasanton, CA) | Prod. #1000212 |
10x Chromium Chip J | 10x Genomics (Pleasanton, CA) | Prod. #1000230 |
NovaSeq S2 Reagent Kit v1.5 | Illumina (San Diego, CA) | Cat. #20028314 |
NuPage 4–12% Bis-Tris Gel | ThermoFisher Scientific (Invitrogen) | Cat. #NP0322 |
PageRuler Pre-stained Protein Ladder, 10–250 kDa | ThermoFisher Scientific | Cat. #26619 |
Revert 700 Total Protein Stain | Li-COR (Lincoln, NE) | Cat. #926-11011 |
SuperSignal West Femto | ThermoFisher Scientific | Cat. #34096 |
Gene Knockout Kit v2 (See Table S3) | Synthego (Redwood, CA) | N/A |
Synthetic sgRNA Kit (See Table S3) | Synthego (Redwood, CA) | N/A |
Neon™ Transfection System 10uL Kit | ThermoFisher Scientific | Cat. #MPK1096 |
Deposited data | ||
Donor 1 uninfected B cell scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695143; SRX13167303 |
Donor 2 uninfected B cell scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695151; SRX13167311 |
Donor 1 B cells day 2 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695145; SRX13167305 |
Donor 2 B cells day 2 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695153; SRX13167313 |
Donor 1 B cells day 5 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695147; SRX13167307 |
Donor 2 B cells day 5 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695155; SRX13167315 |
Donor 1 B cells day 8 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695149; SRX13167309 |
Donor 2 B cells day 8 post-EBV scATAC-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695157; SRX13167317 |
Donor 1 uninfected B cell scRNA-seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695144; SRX13167304 |
Donor 2 uninfected B cell scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695152; SRX13167312 |
Donor 1 B cells day 2 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695146; SRX13167306 |
Donor 2 B cells day 2 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695154; SRX13167314 |
Donor 1 B cells day 5 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695148; SRX13167308 |
Donor 2 B cells day 5 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695156; SRX13167316 |
Donor 1 B cells day 8 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695150; SRX13167310 |
Donor 2 B cells day 8 post-EBV scRNA -seq | This study (GEO accession GSE189141; SRA accession SRP346796) | GSM5695158; SRX13167318 |
GM12878 ATAC-seq | Buenrostro et al., 2013126 (GSE47753) | GSM1155957 |
GM12878 EBNA1 ChIP-seq | Tempera et al., 201679 (GSE73887) | GSM1905009 |
GM12878 EBNALP ChIP-seq | Portal et al., 201380 (GSE49338) | GSM1197603 |
Mutu III EBNA2 ChIP-seq | McLellan et al., 201344 (GSE47629) | GSM1153765 |
Mutu III EBNA3C ChIP-seq | McLellan et al., 201344 (GSE47629) | GSM1153766 |
GM12878 IgG control ChIP-seq | Gunnell et al., 2016127 (GSE76869) | GSM2039169 |
GM12878 RelA ChIP-seq | Zhao et al., 201482; Iannetti et al., 2014 81 (GSE55105) | GSM2628088 |
GM12878 RelB ChIP-seq | Zhao et al., 201482; Iannetti et al., 201481 (GSE55105) | GSM2628090 |
GM12878 cRel ChIP-seq | Zhao et al., 201482; Iannetti et al., 201481 (GSE55105) | GSM2628092 |
GM12878 CTCF ChIP-seq | Lee et al., 2012128 (GSE32883) | GSM489290 |
GM12878 RNA Pol II ChIP-seq | Raha et al., 201085 (GSE19550) | GSM487431 |
GM12878 H2AZ ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733767 |
GM12878 H3K4me1 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733772 |
GM12878 H3K4me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733708 |
GM12878 H3K9ac ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733667 |
GM12878 H3K9me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733664 |
GM12878 H3K27ac ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733771 |
GM12878 H3K27me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733758 |
GM12878 H3K36me3 ChIP-seq | ENCODE Project Consortium 201283 (GSE29611) | GSM733679 |
GM12878 Hi-C | Rao et al., 2014129 (GSE63525) | GSE63525_GM12878_dilution_combined_30.hic |
Oligonucleotides | ||
GAGAAACAUGUCCAUAUAUA | Synthego sgRNA | sgRNA_CD46_1 |
AACUCGUAAGUCCCAUUUGC | Synthego sgRNA | sgRNA_CD46_2 |
UUGCUCCUUAGAGGAAAUAA | Synthego sgRNA | sgRNA_CD46_3 |
GGCCAAGCACGAGAACACUC | Synthego sgRNA | sgRNA_BHLHE40_1 |
CGAGACCACCCGGUGGAGGU | Synthego sgRNA | sgRNA_BHLHE40_2 |
CAGCUCUCCGGCCAAAGGUU | Synthego sgRNA | sgRNA_BHLHE40_3 |
GGAGGUCGAUGUGUUACACC | Synthego sgRNA | sgRNA_MEF2C_1 |
GAGCCAGUGGCAAUAGGUUG | Synthego sgRNA | sgRNA_MEF2C_2 |
GAGACUGGCAUCUCGAAGUU | Synthego sgRNA | sgRNA_MEF2C_3 |
UAGUUGUAACUGAGCGAAAA | Synthego sgRNA | sgRNA_NFE2L2_1 |
GCGACGGAAAGAGUAUGAGC | Synthego sgRNA | sgRNA_NFE2L2_2 |
AUUUGAUUGACAUACUUUGG | Synthego sgRNA | sgRNA_NFE2L2_3 |
UGGGCAGAUCCCGGACCCUU | Synthego sgRNA | sgRNA_JDP2_1 |
CCGAGCUGGGGAGCCCGGUC | Synthego sgRNA | sgRNA_JDP2_2 |
CAAUCAUGGCCCCGAGGUUG | Synthego sgRNA | sgRNA_JDP2_3 |
AAGUAAGUUGAUCUCACCUA | Synthego sgRNA | sgRNA_CD274_dss_1 |
UCCACCAAUUAGAGCACGGC | Synthego sgRNA | sgRNA_CD274_dss_2 |
ACUCAAUGAACCACGAGAGG | Synthego sgRNA | sgRNA_CD274_dss_3 |
UGGGUCUCUGGAAUCACGAC | Synthego sgRNA | sgRNA_CD274_ctrl_1 |
GCGGUGUUGCCCAUGAGAUA | Synthego sgRNA | sgRNA_CD274_ctrl_2 |
TTCAGTGTAATTCTTCTCTGAGCA | IDT | BHLHE40 Forward Primer |
AGGAACGGGGAGCTGATCA | IDT | BHLHE40 Reverse Primer |
AGGGAGGACGAGCAGAAGTA | IDT | MEF2C Forward Primer |
TGGAGTCTAATGAAATGGGGCT | IDT | MEF2C Reverse Primer |
CAGGAGGCTGAGGTTGGAAA | IDT | NFE2L2 Forward Primer |
AACCATTTGTGACTTTGCCCT | IDT | NFE2L2 Reverse Primer |
TGCCTATGATTGGGTCAAGCA | IDT | JDP2 Forward Primer |
GCCCAGTGCTTCTCAAGTGT | IDT | JDP2 Reverse Primer |
Software and algorithms | ||
CellRanger (cellranger-arc) | 10x Genomics (Pleasanton, CA); functions used: cellranger-arc mkref, cellranger-arc mkfastq, cellranger-arc count | v2.0 |
R/R Studio | Open Source/RStudio, PBC (Boston, MA) | v4.0/v1.4.1106 |
Seurat (R package) | Macosko et al., 2015108; Satija et al., 2015109; Stuart et al., 2019110 | v4.0 |
Signac (R package) | Stuart et al.,202134 | v1.4.0 |
Monocle3 (R package) | Qiu et al., 2017130 | v0.2.3 |
Bioconductor (R package) | bioconductor.org; R libraries used include: GenomeInfoDB, ensembldb, EnsDb.Hsapiens.v86, BSgenome.Hsapiens.UCSC.hg38, motifmatcher, JASPAR2020, TFBSTools) | v3.14 |
macs2/macs3 | Liu, 2014113 | v2.2.7/v3.0 |
bedtools | Quinlan and Hall, 2010117; functions used: multiinter | |
GREAT | McLean et al., 201033; http://great.stanford.edu/public/html/ | v4.0.4 |
hicExplorer | Ramirez et al., 2018118; Wolff et al., 2018119; Wolff et al., 2020120 | v3.7.2 |
IGV | Robinson et al., 2011121 | v2.9.2 |
Juicebox | Durand et al.,2016122 | v1.5.1 |
Cytoscape | Shannon et al., 2003123 | v3.8.2 |
Other | ||
RPMI 1640 | Sigma-Aldrich (Raleigh, NC) | Cat. #11875093 |
FBS, heat inactivated | Corning (Durham, NC) | Cat. #35-010-CV |
DMSO | Sigma-Aldrich (Raleigh, NC) | Cat. #D2660-100ML |
10x Chromium Controller | 10x Genomics (Pleasanton, CA) | Prod. #1000204 |
NovaSeq 6000 | Illumina (San Diego, CA) | Cat. #20012850 |
BD FACS Canto II | BD Biosciences (Franklin Lakes, NJ) | N/A |
Sony SH800 Sorter | Sony Biotechnology | N/A |
Neon™ Transfection System | ThermoFisher Scientific | Cat. #MPK5000 |
Human (hg38) reference genome | Genome Reference Consortium | GRCh38.p13 |
Type 1 EBV reference genome | de Jesus et al. 2003131; FASTA accessed via NCBI | NC_007605.1 |
hg38+EBV reference genome | Generated via concatenation of the genomes listed above | N/A |