Although the BCL6 transcriptional repressor is frequently expressed in human follicular lymphomas (FL), its biological role in this disease remains unknown. Herein, we comprehensively identify the set of gene promoters directly targeted by BCL6 in primary human FLs. We noted that BCL6 binds and represses NOTCH2 and NOTCH pathway genes. Moreover, BCL6 and NOTCH2 pathway gene expression is inversely correlated in FL. Notably, BCL6 upregulation is associated with repression of NOTCH2 and its target genes in primary human and murine germinal center (GC) cells. Repression of NOTCH2 is an essential function of BCL6 in FL and GC B cells because inducible expression of Notch2 abrogated GC formation in mice and killed FL cells. Indeed, BCL6-targeting compounds or gene silencing leads to the induction of NOTCH2 activity and compromises survival of FL cells, whereas NOTCH2 depletion or pathway antagonists rescue FL cells from such effects. Moreover, BCL6 inhibitors induced NOTCH2 expression and suppressed growth of human FL xenografts in vivo and primary human FL specimens ex vivo. These studies suggest that established FLs are thus dependent on BCL6 through its suppression of NOTCH2.

Significance: We show that human FLs are dependent on BCL6, and primary human FLs can be killed using specific BCL6 inhibitors. Integrative genomics and functional studies of BCL6 in primary FL cells point toward a novel mechanism whereby BCL6 repression of NOTCH2 drives the survival and growth of FL cells as well as GC B cells, which are the FL cell of origin. Cancer Discov; 7(5); 506–21. ©2017 AACR.

This article is highlighted in the In This Issue feature, p. 443

Follicular lymphoma (FL) is the second most common form of non-Hodgkin lymphoma (1). It is generally an indolent and slow-growing disease that is nonetheless mostly incurable with currently available chemo-immunotherapy regimens (1). FLs arise from germinal center (GC) B cells. GCs are transient structures that develop after exposure to T cell–dependent antigen. To form GCs, B cells aggregate within lymphoid follicles and initiate a program of rapid proliferation and somatic hypermutation of their immunoglobulin loci for the purpose of generating high-affinity antibodies. These rapidly proliferating B cells are called centroblasts and are dependent on the presence of the transcriptional repressor BCL6, which is a master regulator of the GC reaction. As clones of B cells emerge within the GC, they encounter T cells and follicular dendritic cells. Signaling events that ensue select B cells with high-affinity antibody for terminal differentiation into memory and B cells. During this signaling process, the B cells slow down and adopt an intermediate state between pre- and post-GC phenotypes, where they are called centrocytes. FL phenotype and gene expression profiles reflect aspects of centroblasts and centrocytes of the GC reaction.

From the molecular standpoint, FLs almost universally harbor t(14;18) translocations involving fusion of BCL2 to regulatory elements associated with immunoglobulin heavy chain locus (2). Constitutive expression of BCL2 suppresses apoptosis, which would otherwise occur physiologically in GC B cells. Mice engineered to express Bcl2 under the control of the Vav2 promoter develop an FL-like disease, albeit with a long latency period (3). BCL2 is a direct transcriptional target of BCL6, which causes its expression to be completely silenced during the GC reaction. Translocation of BCL2 enables its escape from BCL6 repression. This leads to a situation where both proteins BCL2 and BCL6 are expressed together. Along these lines, it has been reported that >90% of FL cases express BCL6 (4, 5). The implication of BCL6 expression in FL has not been explored.

In normal GC B cells, the most established function of BCL6 is to repress critical checkpoint and DNA-damage repair pathway genes, such as ATR, CHEK1, TP53, CDKN1A, etc. Through this mechanism, GC B cells can proliferate and tolerate the DNA damage associated with somatic hypermutation and class switch recombination (6). The survival and growth checkpoint functions of BCL6 are also maintained in diffuse large B-cell lymphomas (DLBCL), which like FL derive from GC B cells. BCL6 expression in DLBCL is maintained in part through chromosomal translocations, although most DLBCLs express BCL6 regardless of genetic lesions. Functional assays demonstrate that DLBCL cells are dependent on BCL6 regardless of translocations (6). Hence, BCL6 more than an oncogene is actually a lineage factor for DLBCL. BCL6 is a member of the BTB–POZ family of transcription factors and mediates transcriptional repression in large part by recruiting the SMRT, NCOR, and BCOR corepressors via the BTB domain (6). Specific peptidomimetic inhibitors of the BCL6 BTB domain kill DLBCL cells in vitro and in vivo (7–9).

Traditionally, BCL6 has not been considered a phenotypic driver in FL, because these tumors, particularly the low-grade ones, only rarely display BCL6 translocations in their early stages and have an indolent phenotype. However, the potent oncogenic functions of BCL6 make it unlikely that its constitutive expression in FL is merely a passenger marker. BCL6 biological functions are dependent on the target genes that it regulates. The biological functions of BCL6 are not likely limited to repressing cell growth and DNA-damage checkpoints. It is entirely possible that other sets of target genes might be crucial for putative roles of BCL6 in FL. Indeed, previous work showed that BCL6 may function through partially different target genes in DLBCL as compared with normal GC B cells (10). Based on these considerations, we hypothesized that BCL6 might also function as an oncoprotein in FL and that any such role would be linked to repression of specific sets of target genes. Discovery of BCL6 target genes in FL seemed like an appropriate starting point to address these questions. Through this approach, we report a novel function for BCL6 in binding and repressing expression and activity of NOTCH2 in FL cells. Repression of NOTCH2 by BCL6 is required to maintain the survival of FL cells. We show that this function is inherited from GC B cells and is required for development of GCs during the humoral immune response. Finally, we find that BCL6-targeted therapy potently kills FL-derived cell lines both in vitro and in vivo and, most importantly, also kills primary human FL patient specimens ex vivo.

BCL6 Regulates Specific Genes and Pathways in FL, Including NOTCH2

As a first approach to exploring BCL6 functions in FL, we performed chromatin immunoprecipitation (ChIP)-on-chip to identify direct target genes relevant to this disease. Because no cell lines are available that reflect FL biology in its indolent phase, we performed these studies using CD20-purified B cells from four independent primary FL patient samples with >80% tumor cell content. ChIP products were cohybridized with their respective inputs to microarrays representing 25,000 promoters. BCL6 binding sites were identified by random permutation analysis and a peak overlap algorithm (10). BCL6 binding sites (48.4%) overlapped between the four FL specimens, amounting to a total of 1,529 probesets and corresponding to 1,712 genes (Supplementary Table S1). DNA motif analysis confirmed that BCL6 canonical DNA binding sequence was highly enriched at these BCL6 binding sites (P < 1.7−7, FIRE algorithm with hypergeometric test, ref. 11; data not shown). To determine whether BCL6 targets in FL lymphoma cells were associated with particular biological functions, we queried curated gene signatures relevant to lymphomagenesis (12). The top 5 gene sets captured by this method using Fisher exact tests with Benjamini–Hochberg (BH) correction included known direct BCL6 targets from experiments in the Ramos cell line (9), a DLBCL proliferation signature (13), a cell-cycle gene set (14), a NOTCH-induced gene signature (15), and genes repressed by BLIMP1 (ref. 16; Fig. 1A and Supplementary Table S2). We noted that repression of NOTCH was not a previously recognized function of BCL6 in the context of B-cell lymphomas.

Figure 1.

BCL6 displays a specific genomic localization pattern in FL. A, The relative enrichment of specific gene signatures on FL BCL6 target gene sets summarized in a heat map. The statistical significance (BH-adjusted P values) is provided in color key. B, A heat map representation of the relative transcript abundance of BCL6 target genes in FLs that display inverse correlation (P < 0.05, Spearman correlation) with BCL6 expression, from a publicly available dataset of 191 primary FL expression profiles. The color key indicates the relative expression values. Highlighted target genes correspond to the genes found on DAVID analysis; for more details, see Supplementary Fig. S1B. C, Primary FL gene expression profiles were sorted by BCL6 expression from low to high (top row of heat map), and the relative expression values of a set of NOTCH complex and target genes displayed in subsequent rows, indicating their degree of inverse correlation (P values are all <0.05, Spearman correlation) with BCL6. Details are provided in Supplementary Table S4. D, BCL6 binding represented for NOTCH2 and HPRT genes (negative control), in red binding of BCL6 on 4 independent FL patient samples. The y-axis represents read densities normalized to the total number of reads. Threshold setting is explained in the Methods section. Promoter expands to −1,000 base pairs (bp) downstream of transcription start site. E, Cartoon representation of the RBPJ, HES1, MAML1, MAML2, and NOTCH2 promoter regions indicating BCL6 DNA binding motifs (orange dots) and QChIP amplicon location (arrows). F, QChIP assays were performed in DoHH2 and Sc-1 FL cells using BCL6 antibody (black bars) and IgG (negative control, gray bars) for the genes shown in B and a negative control (NEG). The x-axis represents percent enrichment of BCL6 antibody versus input DNA. See additional data in Supplementary Fig. S1.

Figure 1.

BCL6 displays a specific genomic localization pattern in FL. A, The relative enrichment of specific gene signatures on FL BCL6 target gene sets summarized in a heat map. The statistical significance (BH-adjusted P values) is provided in color key. B, A heat map representation of the relative transcript abundance of BCL6 target genes in FLs that display inverse correlation (P < 0.05, Spearman correlation) with BCL6 expression, from a publicly available dataset of 191 primary FL expression profiles. The color key indicates the relative expression values. Highlighted target genes correspond to the genes found on DAVID analysis; for more details, see Supplementary Fig. S1B. C, Primary FL gene expression profiles were sorted by BCL6 expression from low to high (top row of heat map), and the relative expression values of a set of NOTCH complex and target genes displayed in subsequent rows, indicating their degree of inverse correlation (P values are all <0.05, Spearman correlation) with BCL6. Details are provided in Supplementary Table S4. D, BCL6 binding represented for NOTCH2 and HPRT genes (negative control), in red binding of BCL6 on 4 independent FL patient samples. The y-axis represents read densities normalized to the total number of reads. Threshold setting is explained in the Methods section. Promoter expands to −1,000 base pairs (bp) downstream of transcription start site. E, Cartoon representation of the RBPJ, HES1, MAML1, MAML2, and NOTCH2 promoter regions indicating BCL6 DNA binding motifs (orange dots) and QChIP amplicon location (arrows). F, QChIP assays were performed in DoHH2 and Sc-1 FL cells using BCL6 antibody (black bars) and IgG (negative control, gray bars) for the genes shown in B and a negative control (NEG). The x-axis represents percent enrichment of BCL6 antibody versus input DNA. See additional data in Supplementary Fig. S1.

Close modal

To distinguish BCL6 target genes likely to contribute to the FL phenotype, we sought to identify those targets most strongly repressed in FL. Analysis of gene expression profiles from 191 patients with FL (17) demonstrated that 184 FL BCL6 target genes displayed significant inverse correlation with BCL6 expression, including NOTCH2 (Spearman correlation, P < 0.05, Fig. 1B and Supplementary Table S3). To determine whether these 184 genes were enriched for any particular pathway category, we explored their functional annotation using the Database for Annotation, Visualization and Integrated Discovery (DAVID; Supplementary Fig. S1A). This analysis again highlighted NOTCH2 as well as NOTCH pathway genes involved in cell cycle, apoptosis, cellular morphogenesis, lymphoid organ development, or transcription (Supplementary Fig. S1B). These data suggested that BCL6 might be a repressor of the NOTCH2 and NOTCH signaling pathways. In further support of this notion, we observed an inverse correlation between expression of BCL6 and expression of a curated list (15, 18, 19) of NOTCH cofactors and target genes among which NOTCH2 was the most inversely correlated (Spearman correlation, P < 0.05, Fig. 1C and Supplementary Table S4). Examination of BCL6 read densities at the NOTCH2 promoter in the four FL specimens showed enrichment as compared with negative control genes (HPRT and COX6B; Fig. 1D and Supplementary Fig. S1C), similar in magnitude to its enrichment at canonical BCL6 targets like TP53 and BCL6 itself (Supplementary Fig. S1C). Moreover, we identified canonical BCL6 DNA binding sites in the regulatory regions of the NOTCH cofactor genes MAML1, MAML2, and RBPJ, all members of the NOTCH coactivator complex, as well as HES1, a transcriptional repressor and the prototypic NOTCH pathway transcriptional target (Fig. 1E). To validate whether these are true BCL6 targets, we performed QChIP and confirmed that BCL6 is indeed bound to these loci in two independent FL-derived cell lines (Fig. 1F). Primers used to this analysis are found in Supplementary Table S5.

Because GC B cells are the cell of origin of FL, and FL gene expression profiles reflect GC B-cell transcriptional programming we wondered if BCL6 could bind to the NOTCH2 locus in this setting as well. Binding of BCL6 to the promoters of NOTCH2, MAML2, and RBPJ was confirmed by performing QChIP in independent primary human GC B cells (Supplementary Fig. S1D). In addition to FL, GC B cells give rise to DLBCLs. To determine whether BCL6 could bind and regulate NOTCH2 and related genes in DLBCLs, we used BCL6 ChIP-on-chip data performed and analyzed on the same platform as the FLs (10). Among BCL6 target genes in DLBCL cells, the NOTCH-induced gene signature was not significantly enriched (Supplementary Tables S6–S8), although other gene sets overrepresented in FL BCL6 target genes were also enriched in DLBCL (Supplementary Fig. S1E and Supplementary Table S6). Analysis of gene expression profiles from 71 DLBCL patient samples (20) showed that 245 DLBCL BCL6 target genes displayed significant inverse correlation with BCL6 expression (Spearman correlation, P < 0.05; Supplementary Table S7). Nonetheless, analysis of the curated list of NOTCH cofactors and target genes from Fig. 1C indicated that NOTCH2 and other NOTCH pathway partners were inversely correlated (Spearman correlation, P < 0.05; Supplementary Fig. S1G and Supplementary Table S8). Furthermore, less than 15% of the genes found in the DLBCL subset were shared with the ones found on the FL subset (Spearman correlation, P < 0.05; Supplementary Table S9). Altogether, these results point to NOTCH2 and its cofactors as bona fide BCL6 target genes with potential relevance to the phenotype of FL tumors as well as formation of GCs during the humoral immune response. Although repression of the NOTCH pathway is not as strongly linked with DLBCL, it is evident that BCL6 represses NOTCH2 in this disease subtype as well.

BCL6 and NOTCH2 Are Inversely Correlated in GC B Cells

Upregulation of BCL6 is required for mature follicular B cells to differentiate into GC B cells during the humoral immune response. In contrast, NOTCH2 plays a critical role in marginal zone differentiation, which is an alternative cell fate for follicular B cells (21). Hence, we wondered whether upregulation of BCL6 in human GC B cells would be associated with silencing of NOTCH2. We purified primary human naïve B cells (NB) and GC B cells from human tonsils and confirmed their purity by IgD+ and CD38+ staining, respectively (Supplementary Fig. S2A). We then measured the relative transcript abundance of NOTCH2, MAML1, and MAML2 as well as BCL6 by qPCR (Fig. 2A). Whereas BCL6 was upregulated in GC B cells, NOTCH2, MAML1, and MAML2 (but not RBPJ) were concordantly downregulated (Fig. 2A). Examination of available gene expression profiles obtained from five independent sets of NB and GC B cells (22) confirmed downregulation of NOTCH2, MAML1, and NOTCH target genes in GC B cells and the inverse correlation with BCL6 (Fig. 2B and Supplementary Table S10; probesets for MAML2 were not present on this array). NOTCH2 protein downregulation in GC B cells was further confirmed by immunoblotting (Supplementary Fig. S2B). During the GC reaction, B cells first become proliferative centroblasts (CB) and then become centrocytes (CC) as they interact with T cells in the GC light zone. We analyzed gene expression in these cell types using RNA sequencing (RNA-seq), and again observed inverse correlation between BCL6 and NOTCH2 in both CB and CC (Fig. 2C and D).

Figure 2.

Inverse correlation between BCL6 and NOTCH2 complex genes in primary GC B cells. A, qPCR was performed in purified human tonsillar naïve B cells (black) and GC B cells (gray) to measure the relative transcript abundance of the indicated genes. The y-axis represents mRNA expression levels normalized to HPRT. B, A heat map representation of BCL6, NOTCH2, and NOTCH complex and target gene expression levels in five human naïve B cells and five GC B-cell specimens. The color key shows relative expression values. C, Expression values (FPKM) of BCL6 from naïve B cells (purple, n = 5), centroblast (yellow, n = 7) and centrocytes (orange, n = 7) from independent specimens of each. D, Expression values of NOTCH2 as in C. E, Human naïve B cells were cultured with OP9 stromal monolayer and stimulated with IL4 plus IL21 or left untreated. qPCR was performed for the indicated genes. The y-axis represents the fold change, normalized to HPRT, and relative to vehicle (control) at day 4 when maximum levels of BCL6 were reached. F, Mouse resting B220+ cells were isolated and activated with mouse cytokines IL4 and IL21 for 24 hours. The same rationale as for D was followed. For A, E, and F, the mean of three independent experiments is represented along with the SEM. For E and F,P values are based on an unpaired two-tailed t test. *, P < 0.05; **, P < 0.005; ***, P < 0.0001; ns, not significant. See Supplementary Fig. S2 for additional information.

Figure 2.

Inverse correlation between BCL6 and NOTCH2 complex genes in primary GC B cells. A, qPCR was performed in purified human tonsillar naïve B cells (black) and GC B cells (gray) to measure the relative transcript abundance of the indicated genes. The y-axis represents mRNA expression levels normalized to HPRT. B, A heat map representation of BCL6, NOTCH2, and NOTCH complex and target gene expression levels in five human naïve B cells and five GC B-cell specimens. The color key shows relative expression values. C, Expression values (FPKM) of BCL6 from naïve B cells (purple, n = 5), centroblast (yellow, n = 7) and centrocytes (orange, n = 7) from independent specimens of each. D, Expression values of NOTCH2 as in C. E, Human naïve B cells were cultured with OP9 stromal monolayer and stimulated with IL4 plus IL21 or left untreated. qPCR was performed for the indicated genes. The y-axis represents the fold change, normalized to HPRT, and relative to vehicle (control) at day 4 when maximum levels of BCL6 were reached. F, Mouse resting B220+ cells were isolated and activated with mouse cytokines IL4 and IL21 for 24 hours. The same rationale as for D was followed. For A, E, and F, the mean of three independent experiments is represented along with the SEM. For E and F,P values are based on an unpaired two-tailed t test. *, P < 0.05; **, P < 0.005; ***, P < 0.0001; ns, not significant. See Supplementary Fig. S2 for additional information.

Close modal

In order to determine whether these changes in gene expression are linked to GC activation signals, we purified human and murine mature B cells, independently cocultured them with stromal cells (OP9) and exposed them to IL4 and IL21 (23, 24). Murine mature B-cell purity was confirmed by CD45/B220+ staining by flow cytometry (Supplementary Fig. S2C). In both human and murine cells, we observed significant BCL6 upregulation (P < 0.0001; P = 0.0004 human and murine cells, respectively) associated with downregulation of NOTCH2 (P = 0.0136 and 0.044) and MAML2 (P = 0.0029 and 0.0255), although expression of MAML1 (P = 0.1069 and 0.0609) and RBPJ (P = 0.0784 and 0.1841) was more variable (Fig. 2E and F). These data suggest that BCL6 repression of NOTCH2 is an integral feature of normal GC B-cell activation and may be critical to specifying the GC phenotype in opposition to marginal zone differentiation.

NOTCH2 Expression Impairs GC Formation

Given that BCL6 is required for the development of GC formation and directly represses NOTCH2, we wondered whether expression of an active form of NOTCH2 (intracellular domain, ICN2) in GC B cells might disrupt GC formation. To address this question, we studied GC formation in a mouse strain engineered to contain an ICN2-IRES-YFP (ICN2) cassette with a LoxP flanked start site knocked-in to the ROSA26 locus (25). These mice were crossed with a tamoxifen-inducible ROSA26-Cre-ERT2 strain or in ROSA26–wild-type (WT) control mice. GC formation was induced by immunization with the T cell–dependent antigen NP65-CGG. ICN2 expression was induced the following day by tamoxifen injection. Animals were sacrificed 14 days later, and spleens were resected for analysis (Supplementary Fig. S3A). Paraffin-embedded spleen sections from WT and ICN2 mice were stained for the GC-specific marker peanut agglutinin (PNA) or for BCL6 and B220. GCs were defined as clusters of PNA+ or BCL6+/B220+ cells (Fig. 3A and B). As compared with WT, ICN2 conditional mice exhibited significant reduction in PNA+ or BCL6+/B220+ GC per spleen section (mean of 29 vs. 4, P = 0.0005, unpaired two-tailed t test; and mean 22 vs. 4 P = 0.0003, respectively; Fig. 3C and D). The average size of the GCs was also reduced by 2- to 3-fold versus WT controls (mean of 262 ± 36 vs. 91 ± 10 μm2 for PNA+, P = 0.0015, and 177 ± 30 vs. 84 ± 13 μm2 for BCL6+/B220+, P = 0.0336 respectively; Fig. 3E–G). A more quantitative analysis of GC B cells was generated by flow cytometry. In the presence of ICN2, the abundance of B220+GL7+CD95+ GC B cells was reduced 3-fold as compared with WT animals (1.7% vs. 0.6% mean GC B cells vs. total splenocytes, P = 0.0003 unpaired two-tailed t test; Fig. 3H and I).

Figure 3.

GC reaction is impaired in NOTCH2 knock-in mice. A, Representative images of spleen sections from WT control and ICN2 knock-in mice stained for GC marker PNA 14 days after immunization with NP65-CGG. The black squares on left column (4×) highlight GCs, which are shown at 20× amplification in the right column. Scale bars, 100 μm (4×) and 20 μm (20×). B, Same experiment performed in panel A for BCL6+/B220+ staining. C, The number of GCs per spleen (y-axis) from immunohistochemistry for PNA+ clusters shown in A. D, The number of GCs per spleen (y-axis) from immunohistochemistry of BCL6+B220+ clusters shown in B. C and D, The range bars represent the mean values and SEM and the P values are shown on top. E, The surface area occupied by GCs in the spleens of immunized and induced ICN2 and control mice is shown for PNA staining and is represented by their area in μm2 (y-axis). F, The same as in panel E for BCL6/B220 staining. E and F, The average of the means for each group is shown. Each column corresponds to an individual mouse; each point is an individual GC; P values are shown on top. SEM and P values are shown. G, Average of GC area (μm2) of WT (black) and ICN2 (green) mice from IHC of paraffin-embedded spleen slides stained with PNA+ and BCL6+/B220+ antibodies. The mean values are 262 ± 36 versus 91 ± 10 μm2 for PNA+ GC, and 177 ± 30 versus 84 ± 13 μm2 for BCL6+/B220+. Data are shown for immunized CγCre-WT (n = 5) and CγCre-ICN2 (n = 8) mice. The statistical significance of this difference is shown based on unpaired two-tailed t test (P = 0.0015 and P = 0.0336, respectively). H, Flow-cytometry analysis of B220+GL7+CD95+ labeled splenic GC B-cell populations. Cells were gated for B220+, GL7 is on the y-axis and CD95(Fas) is on the x-axis. The percentage of double positive cells corresponds to GC B cells (black box). I, The percentage of B220+ gated GL7+/CD95+ GC B cells among total splenocytes is shown from the spleens of immunized WT (n = 5) and ICN2 induced (n = 8) mice. In the presence of ICN2, the abundance of B220+GL7+CD95+ GC B cells was reduced 3-fold as compared with WT animals (1.7% vs. 0.6% mean GC B cells vs. total splenocytes, P = 0.0003 unpaired two-tailed t test). See Supplementary Figs. S3 and S4 for additional data.

Figure 3.

GC reaction is impaired in NOTCH2 knock-in mice. A, Representative images of spleen sections from WT control and ICN2 knock-in mice stained for GC marker PNA 14 days after immunization with NP65-CGG. The black squares on left column (4×) highlight GCs, which are shown at 20× amplification in the right column. Scale bars, 100 μm (4×) and 20 μm (20×). B, Same experiment performed in panel A for BCL6+/B220+ staining. C, The number of GCs per spleen (y-axis) from immunohistochemistry for PNA+ clusters shown in A. D, The number of GCs per spleen (y-axis) from immunohistochemistry of BCL6+B220+ clusters shown in B. C and D, The range bars represent the mean values and SEM and the P values are shown on top. E, The surface area occupied by GCs in the spleens of immunized and induced ICN2 and control mice is shown for PNA staining and is represented by their area in μm2 (y-axis). F, The same as in panel E for BCL6/B220 staining. E and F, The average of the means for each group is shown. Each column corresponds to an individual mouse; each point is an individual GC; P values are shown on top. SEM and P values are shown. G, Average of GC area (μm2) of WT (black) and ICN2 (green) mice from IHC of paraffin-embedded spleen slides stained with PNA+ and BCL6+/B220+ antibodies. The mean values are 262 ± 36 versus 91 ± 10 μm2 for PNA+ GC, and 177 ± 30 versus 84 ± 13 μm2 for BCL6+/B220+. Data are shown for immunized CγCre-WT (n = 5) and CγCre-ICN2 (n = 8) mice. The statistical significance of this difference is shown based on unpaired two-tailed t test (P = 0.0015 and P = 0.0336, respectively). H, Flow-cytometry analysis of B220+GL7+CD95+ labeled splenic GC B-cell populations. Cells were gated for B220+, GL7 is on the y-axis and CD95(Fas) is on the x-axis. The percentage of double positive cells corresponds to GC B cells (black box). I, The percentage of B220+ gated GL7+/CD95+ GC B cells among total splenocytes is shown from the spleens of immunized WT (n = 5) and ICN2 induced (n = 8) mice. In the presence of ICN2, the abundance of B220+GL7+CD95+ GC B cells was reduced 3-fold as compared with WT animals (1.7% vs. 0.6% mean GC B cells vs. total splenocytes, P = 0.0003 unpaired two-tailed t test). See Supplementary Figs. S3 and S4 for additional data.

Close modal

Formation of GCs requires cooperation between different cell types. Hence, it is conceivable that ER-induced ICN2 impairment in GC formation could be attributed to non–B-cell effects. Therefore, we established a second mouse model that specifically limited conditional expression of ICN2 to GC B cells. In this case, the C1γCre mouse strain, which activates Cre expression in GC B cells (26), was crossed to ROSA26-WT or ROSA26-ICN2-IRES-YFP (hereafter referred to as WT and ICN2, respectively). These animals were immunized with the T cell–dependent antigen sheep red blood cells (SRBC). Paraffin-embedded spleen sections from WT and ICN2 mice were stained for PNA, or BCL6/B220, to identify GCs (Supplementary Fig. S3B and S3C). These experiments again yielded significant reductions in the numbers of GCs in ICN2 conditional mice, with a mean of 13 versus 4 PNA+ GC per spleen section (P = 0.0001, unpaired two-tailed t test) and 14 versus 5 BCL6+/B220+ GC per spleen (P = 0.0001; Supplementary Fig. S3D and S3E). The average size of the GCs was also significantly diminished in ICN2-expressing mice (mean of 301 ± 35 vs. 68 ± 17 μm2 for PNA+, P < 0.0001, and 226 ± 24 vs. 66 ± 16 μm2 for BCL6+/B220+, P = 0.0002 respectively; Supplementary Fig. S3F–S3H). Quantitative assessment of GC B cells by flow cytometry yielded a 3-fold reduction in the abundance of B220+GL7+CD95+ GC B cells as compared with WT animals (3.28% vs. 1.08% mean GC B cells vs. total splenocytes, P < 0.0001, unpaired two-tailed t test; Supplementary Fig. S4A and S4B).

Consistent with the reduction in GCs (which contain cells that proliferate and undergo apoptosis), there was also reduction in the abundance of proliferating cell clusters as shown by PCNA and Ki67 immunohistochemistry (Supplementary Fig. S4C), as well as clusters of cells with apoptotic markers caspase-3 and TUNEL in the ICN2 mice (Supplementary Fig. S4D). Given the crucial role of ICN2 in driving marginal zone B-cell differentiation, we next stained for MZB markers CD21 and CD23. We observed an increase in the MZB cell population in ICN2 conditional mice in detriment to the follicular B (FoB) cell population (mean of 65% ± 1.5% FoB vs. 15% ± 2.6% MZB) compared with WT (mean of 70% ± 3% FoB vs. 10% ± 1.3% MZB; Supplementary Fig. S4E). In contrast, as expected, there was no effect on T lymphoid (single- or double-positive populations stained for CD8/CD4; top) or myeloid lineages (CD11b/Gr-1; bottom, Supplementary Fig. S4F). Altogether, these data indicate that ICN2 expression is incompatible with B cells forming GCs and that repression of Notch2 is a critical function of BCL6 in enabling the GC phenotype.

BCL6 Represses NOTCH2 Expression and Activity in FL Cells.

To confirm that BCL6 directly represses NOTCH2 and related genes, we depleted BCL6 from FL-derived cell lines using an siRNA that we validated to be specific for BCL6 (27) compared with scrambled control, followed by qPCR assessment of NOTCH2, MAML1, MAML2, and RBPJ transcripts (Fig. 4A). We observed approximately 2-fold derepression of NOTCH2 and variable derepression of the other genes. This magnitude of derepression is similar to that reported for other BCL6 targets (7, 9, 10, 28, 29). siBCL6 was confirmed to deplete BCL6 protein (Supplementary Fig. S5A). To further confirm this result using an independent approach, we treated the FL cell lines with the specific BCL6 inhibitor RI-BPI, which binds to the BCL6 BTB repression domain to block the transcriptional effects of BCL6 (7). We first validated that we could reproduce the known effect of Retro-inverted BCL6-peptide inhibitor (RI-BPI) in blocking the repressor activity of the BCL6 BTB domain in the context of lymphoma cell lines using a BCL6 BTB domain reporter assay (Supplementary Fig. S5B). We then measured the effect of RI-BPI on derepressing NOTCH2, MAML1, MAML2, and RBPJ as compared with control peptide and observed a similar effect as that seen with siRNA (Fig. 4B). There was no induction of NOTCH1, which was expressed at very low levels in these cells (data not shown). Given that NOTCH2 was also inversely correlated with BCL6 in GCB-DLBCL cells, we examined the effect of RI-BPI on two such cell lines and observed a generally similar degree of derepression of NOTCH2, MAML1, and MAML2 (Supplementary Fig. S5C). To determine whether upregulation of NOTCH2 was functionally significant, we performed NOTCH reporter assays in the FL-derived cell lines after BCL6 siRNA. BCL6 knockdown resulted in significant induction of NOTCH reporter activity (P < 0.0001 in both cell lines, unpaired two-tailed t test), but did not affect a control reporter (Fig. 4C).

Figure 4.

BCL6 represses NOTCH2 complex genes and NOTCH activity. A, The relative transcript abundance of NOTCH2, MAML1, MAML2, and RBPJ was examined by qPCR in DoHH2 (black bars) and Sc-1 (gray bars) 72 hours after BCL6 siRNA depletion versus control siRNA. Values were normalized to HPRT, and fold change (y-axis) is represented over a scrambled siRNA control. B, The relative transcript abundance of NOTCH2, MAML1, MAML2, and RBPJ was examined by qPCR in FL cell lines DoHH2 (black bars) and Sc-1 (gray bars) 72 hours after RI-BPI treatment (15 μmol/L RI-BPI) on the right. Values were normalized to HPRT, and fold change (y-axis) is represented over control peptide (CP). C, Reporter assays performed in DoHH2 (black bars) and Sc-1 (gray bars) cells transfected with pGL2-HESAB (NOTCH reporter) or pGL2 control vector, and with BCL6 (siBCL6) or control siRNA (siC). The y-axis shows the luciferase activity relative to renilla (internal control). All panels represent the mean of three independent experiments, each performed in triplicate; error bars, SEM. The statistical values are based on an unpaired two-tailed t test. ***, P < 0.0001. D, Expression values (FPKM) of NOTCH ligands DLL1, DLL3, DLL4, JAG1, and JAG2 on DoHH2 (blue), Sc-1 (yellow), OCI-Ly1 (purple), SU-DHL-4 (green). E, As in D, expression values (FPKM) of metalloproteases ADAM10 and ADAM17 on the same cell lines. F, Expression values (FPKM) of DLL1 (top), DLL3 (middle), and DLL4 (bottom) from left to right: naïve B cells (NB; n = 5), GC B cells (GCB; n = 4), centroblast (CB; n = 7), centrocytes (CC; n = 0.7), bone marrow plasma cells (BMPC; n = 3), tonsillar plasma cells (TPC; n = 5), memory B cells (MB; n = 8), and FL (n = 77) from independent specimens of each. *, P ≤ 0.05. Square dots are outliers (below the first quartile or above the fourth quartile). G, As in F, expression values (FPKM) of JAG1 (top) and JAG2 (bottom). H, As in G, expression values (FPKM) of ADAM10 (top) and ADAM 17 (bottom). I, Flow cytometry to assess apoptosis of FL cell lines driven by DLL1 ligand. The percentage of apoptotic cells is observed on the upper-right quadrant of double-positive labeled cells for propidium iodide (PI; y-axis) and Annexin V (x-axis) on DoHH2 (top) and Sc-1 (bottom) cell lines cocultured with HS5-control (right graph) or HS5-DLL1 (triplicate in left graphs) stromal cell line for 48 hours. See Supplementary Fig. S5 for additional data.

Figure 4.

BCL6 represses NOTCH2 complex genes and NOTCH activity. A, The relative transcript abundance of NOTCH2, MAML1, MAML2, and RBPJ was examined by qPCR in DoHH2 (black bars) and Sc-1 (gray bars) 72 hours after BCL6 siRNA depletion versus control siRNA. Values were normalized to HPRT, and fold change (y-axis) is represented over a scrambled siRNA control. B, The relative transcript abundance of NOTCH2, MAML1, MAML2, and RBPJ was examined by qPCR in FL cell lines DoHH2 (black bars) and Sc-1 (gray bars) 72 hours after RI-BPI treatment (15 μmol/L RI-BPI) on the right. Values were normalized to HPRT, and fold change (y-axis) is represented over control peptide (CP). C, Reporter assays performed in DoHH2 (black bars) and Sc-1 (gray bars) cells transfected with pGL2-HESAB (NOTCH reporter) or pGL2 control vector, and with BCL6 (siBCL6) or control siRNA (siC). The y-axis shows the luciferase activity relative to renilla (internal control). All panels represent the mean of three independent experiments, each performed in triplicate; error bars, SEM. The statistical values are based on an unpaired two-tailed t test. ***, P < 0.0001. D, Expression values (FPKM) of NOTCH ligands DLL1, DLL3, DLL4, JAG1, and JAG2 on DoHH2 (blue), Sc-1 (yellow), OCI-Ly1 (purple), SU-DHL-4 (green). E, As in D, expression values (FPKM) of metalloproteases ADAM10 and ADAM17 on the same cell lines. F, Expression values (FPKM) of DLL1 (top), DLL3 (middle), and DLL4 (bottom) from left to right: naïve B cells (NB; n = 5), GC B cells (GCB; n = 4), centroblast (CB; n = 7), centrocytes (CC; n = 0.7), bone marrow plasma cells (BMPC; n = 3), tonsillar plasma cells (TPC; n = 5), memory B cells (MB; n = 8), and FL (n = 77) from independent specimens of each. *, P ≤ 0.05. Square dots are outliers (below the first quartile or above the fourth quartile). G, As in F, expression values (FPKM) of JAG1 (top) and JAG2 (bottom). H, As in G, expression values (FPKM) of ADAM10 (top) and ADAM 17 (bottom). I, Flow cytometry to assess apoptosis of FL cell lines driven by DLL1 ligand. The percentage of apoptotic cells is observed on the upper-right quadrant of double-positive labeled cells for propidium iodide (PI; y-axis) and Annexin V (x-axis) on DoHH2 (top) and Sc-1 (bottom) cell lines cocultured with HS5-control (right graph) or HS5-DLL1 (triplicate in left graphs) stromal cell line for 48 hours. See Supplementary Fig. S5 for additional data.

Close modal

In addition to induction of expression of the NOTCH2 transcriptional complex, NOTCH activation involves signaling through NOTCH ligands (DLL1, DLL3, DLL4, JAG1, and JAG2) and cleavage by metalloproteases (ADAM10 and ADAM17). To determine possible sources of NOTCH signaling in FLs and GC B cells, we measured expression of these genes in the principal FL and DLBCL cell lines used for this study (DoHH2, Sc-1, OCI-Ly1, and SUD-HL-4), purified primary mature B-cell populations: NB, GC, CB, CC, memory B (MB), tonsillar plasma cell (TPC), bone marrow plasma cell (BMPC), and in a cohort of patients with primary FL by RNA-seq (30, 31). Expression of NOTCH ligands DLL1, DLL3, DLL4, and JAG1 was low in all of four cell lines, although JAG2 was expressed, and possibly relevant to NOTCH activation in vitro (Fig. 4D and E). In contrast, NOTCH ligand expression was essentially absent in the relevant mature B-cell subsets, although DLL4 and JAG1 were upregulated later in BMPCs (Fig. 4F and G). Only a small subset of FLs manifested higher levels of NOTCH ligands. In contrast, ADAM10 and ADAM17 are expressed in the cell lines, primary mature B cells and FLs, especially ADAM17 (Fig. 4D and H). Hence, in the in vivo setting, NOTCH ligand delivery to lymphoma cells likely comes mostly from the lymph node microenvironment, where NOTCH ligands are known to be expressed (32), whereupon FLs or mature B cells are then competent to cleave NOTCH2. Indeed, we observed that coculture of FL cells with a stromal cell line engineered to express DLL1 but not the same cell line without DLL1 reproducibly induced apoptosis, consistent with NOTCH2 signaling being deleterious to these cells (Fig. 4I).

BCL6 Maintains the Survival of FL Cells in a NOTCH2-Dependent Manner

To determine whether BCL6 repression of NOTCH2 was important to its actions in FL cells, we first wished to establish whether FL cells are biologically dependent on BCL6. We therefore exposed DoHH2, Sc-1, and WSU-DLCL2 FL-derived cell lines to increasing concentrations of RI-BPI and measured cell viability using a fluorometric resazurin reduction method. DoHH2 and Sc-1 cells displayed a GI50 of 11.7 μmol/L and 15.2 μmol/L, respectively, which is comparable to the GI50 of BCL6-dependent DLBCL cells (7), whereas WSU-DLCL2 cells were more resistant (Fig. 5A). Similar to the case of DLBCL (7, 9), not all FL cells were responsive to RI-BPI, but the ones that were sensitive underwent apoptosis, as shown in caspase-3/7 cleavage assays and annexin V/7AAD flow cytometry (Fig. 5B and Supplementary Fig. S6A). To determine whether NOTCH2 repression contributes to the effect of BCL6 in maintaining survival of FL cells, we attempted to rescue our panel of BCL6-dependent FL and GCB-DLBCL lymphoma cell lines (DoHH2, Sc-1, SU-DHL-4, and OCI-Ly1) from the effects of BCL6 depletion by preventing NOTCH2 upregulation using an siRNA approach. We verified knockdown of both transcripts (BCL6 and NOTCH2) in each cell line, using two independent siRNA for both BCL6 and NOTCH2 (Supplementary Fig. S6B). BCL6 knockdown resulted in a ∼30% to 60% loss of viability in all four cell lines at 48 hours, whereas NOTCH2 siRNA alone did not affect cell viability (Fig. 5C and Supplementary Fig. S6C). Notably, concordant knockdown of NOTCH2 prevented its upregulation in response to BCL6 siRNA and significantly rescued all of four cell lines from BCL6 siRNA–induced loss of viability. We used two independent siRNA sequences for BCL6 and NOTCH2. The rescue of siBCL6-1 sequence by both siNOTCH2 RNA sequences is shown in Fig. 5C and the rescue of siBCL6-2 in Supplementary Fig. S6C. In contrast, the viability of the BCL6-independent t(14;18) lymphoma cell line OCI-Ly8 (8) was not affected by BCL6 or NOTCH2 siRNA, even though they manifested a similar degree of knockdown (Supplementary Fig. S6D and S6E). NOTCH2 siRNA also, at least partially, rescued FL cells from loss of viability induced by the BCL6 inhibitor RI-BPI (Fig. 5D and data not shown). In a reciprocal experiment, to determine whether induction of NOTCH2 was sufficient to suppress the growth of FL cells downstream of BCL6 inhibition, we transduced DoHH2 and Sc-1 cells with a retrovirus expressing ICN2 and GFP, or GFP alone. We measured the relative depletion of GFP-positive cells from the total population of cells by flow cytometry over the course of 10 days. NOTCH2 expression was clearly growth suppressive and sufficient to inhibit FL cells, because 95% of ICN2-GFP DoHH2 cells were depleted by 10 days, as were 75% of ICN2-GFP Sc-1 cells (Supplementary Fig. S6F). Finally, to confirm the importance of NOTCH2 pathway suppression by BCL6 in FL cells, we exposed lymphoma cells to the NOTCH2 antagonist antibody NRR2, after confirming its specificity of action in vivo against NOTCH2 but not NOTCH1, consistent with previous reports (Supplementary Fig. S7; refs. 33, 34). NRR2, but not control antibody, could also rescue DoHH2 and SU-DHL-4 cells from cell death induced by RI-BPI to variable degrees (Fig. 5E). Repression of NOTCH2 is thus a critical downstream mechanism of action of BCL6, required for its ability to maintain the survival of FL cells.

Figure 5.

FL cells are dependent on BCL6 in a NOTCH2-dependent manner. A, DoHH2, Sc-1, and WSU-DLCL2 FL cell lines were exposed to six concentrations of RI-BPI (from 1 to 40 μmol/L) or vehicle (water) for 48 hours. The x-axis shows the dose of RI-BPI. The y-axis shows the fractional effect of RI-BPI versus control on cell viability. The experiment was done in triplicate. The dose (in μmol/L) that inhibited cell growth by 50% (GI50) is shown next to each cell line. B, Luminescent caspase-7 and -3 activity assays were performed in FL cell lines (x-axis) exposed to vehicle (gray columns) or RI-BPI 10 μmol/L (black columns) for 24 hours. Results are expressed in percentage of relative luciferase units (RLU) to control (y-axis). C, DoHH2, Sc-1, SU-DHL-4, and OCI-Ly1 cells were transfected NOTCH2 siRNA, BCL6 siRNA, or both as indicated, and cell viability was measured at 48 and 72 hours. The y-axis represents percentage cell viability, normalized to control siRNA (siC; dotted line). D, DoHH2 cells were transfected with NOTCH2 siRNA as indicated and control siRNA as indicated, 24 hours after transfection cells were treated with 10 μmol/L RI-BPI and 24 hours after treatment cell viability was measured as described before. The figure shows the mean of 3 experiments with SEM. E, DoHH2 and SU-DHL-4 cells were pretreated with anti-IgG1 or NOTCH2 antagonist antibody NRR2 for 24 hours and then exposed to 10 μmol/L RI-BPI or control peptide (CP). The y-axis represents viability (at 24 and 48 hours) relative to control antibody (anti-IgG1) treated with CP. All experiments, unless otherwise indicated, were performed in triplicate; the error bars, SEM. Statistical significance is shown (P value one-tailed t test): *, P < 0.05; **, P < 0.005. See Supplementary Figs. S6 and S7 for additional data.

Figure 5.

FL cells are dependent on BCL6 in a NOTCH2-dependent manner. A, DoHH2, Sc-1, and WSU-DLCL2 FL cell lines were exposed to six concentrations of RI-BPI (from 1 to 40 μmol/L) or vehicle (water) for 48 hours. The x-axis shows the dose of RI-BPI. The y-axis shows the fractional effect of RI-BPI versus control on cell viability. The experiment was done in triplicate. The dose (in μmol/L) that inhibited cell growth by 50% (GI50) is shown next to each cell line. B, Luminescent caspase-7 and -3 activity assays were performed in FL cell lines (x-axis) exposed to vehicle (gray columns) or RI-BPI 10 μmol/L (black columns) for 24 hours. Results are expressed in percentage of relative luciferase units (RLU) to control (y-axis). C, DoHH2, Sc-1, SU-DHL-4, and OCI-Ly1 cells were transfected NOTCH2 siRNA, BCL6 siRNA, or both as indicated, and cell viability was measured at 48 and 72 hours. The y-axis represents percentage cell viability, normalized to control siRNA (siC; dotted line). D, DoHH2 cells were transfected with NOTCH2 siRNA as indicated and control siRNA as indicated, 24 hours after transfection cells were treated with 10 μmol/L RI-BPI and 24 hours after treatment cell viability was measured as described before. The figure shows the mean of 3 experiments with SEM. E, DoHH2 and SU-DHL-4 cells were pretreated with anti-IgG1 or NOTCH2 antagonist antibody NRR2 for 24 hours and then exposed to 10 μmol/L RI-BPI or control peptide (CP). The y-axis represents viability (at 24 and 48 hours) relative to control antibody (anti-IgG1) treated with CP. All experiments, unless otherwise indicated, were performed in triplicate; the error bars, SEM. Statistical significance is shown (P value one-tailed t test): *, P < 0.05; **, P < 0.005. See Supplementary Figs. S6 and S7 for additional data.

Close modal

BCL6 Inhibitors Suppress FL Xenografts In Vivo and Primary Human FLs Ex Vivo

FL cell lines may not necessarily accurately represent the biology of primary indolent FL in human patients at the time of diagnosis. We obtained a set of 17 diagnostic primary human FL specimens from patients with nontransformed disease, made single-cell suspensions, exposed them to RI-BPI or vehicle ex vivo for 48 hours, and then assessed for viability. Immunohistochemistry analysis indicated that 10 patients were clearly BCL6 positive and 7 were borderline positive to negative for BCL6 (data not shown). All 17 samples were exposed to 10 μmol/L RI-BPI or vehicle. Although the BCL6-negative/low FLs were resistant to BCL6 inhibitors, 9 of 10 of the BCL6-positive FLs responded with a 20% to 70% loss of viability (Fig. 6A). Consistent with the actions of BCL6 in normal GC B cells and FL cell lines, we observed that in primary human FLs, RI-BPI induced derepression of NOTCH2, as well as induction of the NOTCH2 targets HES1 and HES6 (Fig. 6B). Moreover, we also observed reexpression of ATF5, APOL6, CCR6, and HOXA13, all of which are direct BCL6 targets inversely correlated with BCL6 expression in patients with FL (Fig. 1B and Supplementary Table S3), and of STAT3, a positive control BCL6 target, but not of HPRT, which is a negative control (Supplementary Fig. S8A).

Figure 6.

RI-BPI suppresses FL tumors in vivo and ex vivo. A, Single-cell suspensions of 17 confirmed FL specimens were exposed to vehicle (control line) or 20 μmol/L of RI-BPI (except case 14 that was treated with 5 μmol/L) for 48 hours. Seven samples were BCL6 negative (gray bars) and 10 samples were BCL6 positive (black bars). Cell viability (represented as a percentage of control-treated cells) is shown on the y-axis. Individual cases as well as the average for all the cases (m) are shown on the x-axis. Statistical significance (unpaired t test) was determined for the average of BCL6-positive versus BCL6-negative cases. The experiment was carried out in duplicate. B, An FL specimen exposed to 10 μmol/L RI-BPI was harvested 48 hours after treatment and mRNA abundance examined by qPCR for NOTCH2, HES6, and HES1 and normalized to HPRT. Results are expressed as fold induction compared with control (vehicle). C, Tumor growth plots in DoHH2 (left) and Sc-1 (right) xenografted mice treated with vehicle (PBS, n = 5, gray lines) or RI-BPI 25 mg/kg/day (n = 5, black lines) for 10 consecutive days. The y-axis indicates tumor volume (in mm3) and x-axis days of treatment. The P values represent the comparison of tumor volumes in treated to control mice at day 10 by the Student t test. D, Representative immunohistochemistry images from DoHH2 and Sc-1 tumors after treatment with control or RI-BPI assayed for apoptosis by TUNEL and caspase-3 staining (top and bottom panels, respectively). Scale bar, 50 μm. E, qPCR was performed in triplicate from the DoHH2 FL xenografts of mice treated with vehicle (n = 4) or RI-BPI 25 mg/kg/day for 7 days (n = 4) to assess transcript abundance of NOTCH2, MAML1, MAML2, and HES1, normalized to HPRT. Statistical significance was determined by the Mann–Whitney test. See Supplementary Fig. S8 for additional data.

Figure 6.

RI-BPI suppresses FL tumors in vivo and ex vivo. A, Single-cell suspensions of 17 confirmed FL specimens were exposed to vehicle (control line) or 20 μmol/L of RI-BPI (except case 14 that was treated with 5 μmol/L) for 48 hours. Seven samples were BCL6 negative (gray bars) and 10 samples were BCL6 positive (black bars). Cell viability (represented as a percentage of control-treated cells) is shown on the y-axis. Individual cases as well as the average for all the cases (m) are shown on the x-axis. Statistical significance (unpaired t test) was determined for the average of BCL6-positive versus BCL6-negative cases. The experiment was carried out in duplicate. B, An FL specimen exposed to 10 μmol/L RI-BPI was harvested 48 hours after treatment and mRNA abundance examined by qPCR for NOTCH2, HES6, and HES1 and normalized to HPRT. Results are expressed as fold induction compared with control (vehicle). C, Tumor growth plots in DoHH2 (left) and Sc-1 (right) xenografted mice treated with vehicle (PBS, n = 5, gray lines) or RI-BPI 25 mg/kg/day (n = 5, black lines) for 10 consecutive days. The y-axis indicates tumor volume (in mm3) and x-axis days of treatment. The P values represent the comparison of tumor volumes in treated to control mice at day 10 by the Student t test. D, Representative immunohistochemistry images from DoHH2 and Sc-1 tumors after treatment with control or RI-BPI assayed for apoptosis by TUNEL and caspase-3 staining (top and bottom panels, respectively). Scale bar, 50 μm. E, qPCR was performed in triplicate from the DoHH2 FL xenografts of mice treated with vehicle (n = 4) or RI-BPI 25 mg/kg/day for 7 days (n = 4) to assess transcript abundance of NOTCH2, MAML1, MAML2, and HES1, normalized to HPRT. Statistical significance was determined by the Mann–Whitney test. See Supplementary Fig. S8 for additional data.

Close modal

To determine whether RI-BPI could also suppress FL tumors in situ in animals, we xenotransplanted the DoHH2 and Sc-1 cell lines into SCID mice. Once palpable tumors formed, pairs of DoHH2 or Sc-1 tumor–bearing mice were randomized to receive either RI-BPI 25 mg/kg/day intraperitoneally or vehicle control (5 mice per treatment condition). Both animals of each pair were sacrificed when one of them reached maximal permitted tumor burden. In all cases, the RI-BPI–treated tumors were considerably smaller than controls for both the more sensitive DoHH2 (Fisher exact test P = 0.03) and the less sensitive Sc-1 cell line (Fisher exact test P = 0.04, Fig. 6C). Immunohistochemical analysis of these tumors showed an increase in apoptosis in the RI-BPI–treated tumors by TUNEL assays from 7% to 12% in DoHH2 (P < 0.001, Fisher exact test), and from 10% to 18% (P < 0.001, Fisher exact test) in Sc-1 tumors, respectively. Analysis of these same tumors by caspase-3 yielded similar results, from 6% to 22% in DoHH2 (P < 0.0001, Fisher exact test), and from 11% to 21% (P < 0.0001, Fisher exact test) in Sc-1 (Fig. 6D and Supplementary Fig. S8B–S8D). Examination of mRNA extracted from tumor xenografts revealed upregulation of NOTCH2, MAML1, MAML2, and HES1 (P = 0.0086, 0.0011, 0.0929, and 0.0079, respectively, Mann–Whitney test) in RI-BPI–treated mice versus vehicle (Fig. 6E). BCL6 is thus a bona fide therapeutic target in FL at least in part through its repression of NOTCH2, which we show is a growth suppressor in FL.

The role of BCL6 in FL has not been previously explored, in part because of considerations such as (i) the indolent phenotype of FL distinct from the more aggressive DLBCLs typically associated with BCL6, (ii) the frequent t(14:18) translocation focused attention on BCL2, and (iii) BCL6 is not often translocated in FL (5). However, our previous work indicated that DLBCL cells are dependent on BCL6 regardless of whether its locus is affected by mutations (7, 8). Therefore, analysis of BCL6 in this disease seemed warranted (35, 36). Analysis of the BCL6 cistrome in primary FL specimens suggested new mechanisms of action for BCL6 not previously gleaned from studies in DLBCL.

In particular, we focused on BCL6 repression of NOTCH2, a growth suppressor of pre–B leukemia cells and Hodgkin lymphoma cells (37). BCL6 was bound to NOTCH target genes in FL, and NOTCH2 levels were inversely correlated with BCL6 in FL. We find that BCL6 is a direct repressor of NOTCH2, MAML1, and MAML2 expression, and that BCL6 inhibition induces NOTCH2 transcriptional activity in reporter assays and endogenous NOTCH2 target genes in FL cells. BCL6 was also shown to antagonize NOTCH signaling in the context of Xenopus embryonic development (38). In that setting, BCL6 binding and repression of certain NOTCH target genes as well as its direct interference in the NOTCH1–MAML1 interaction was essential in the determination of axis symmetry during development (38). In mammals, BCL6 does not play a role in axis symmetry because Bcl6-null mice do not show this developmental defect, and antagonism with NOTCH signaling is more linked to NOTCH2, at least in B cells. More recently, it was also reported that BCL6 may control neurogenesis through SIRT1-dependent repression of selective NOTCH targets (39). BCL6 prevented transcriptional activation of the Hes5 promoter, by excluding Maml1 and instead recruiting Sirt1 to NOTCH transcriptional complex to downregulate its expression but without impairing NOTCH signaling during the transition from neuronal progenitor stem cells to differentiated neurons (39). This additional protein interference mechanism may be relevant to B cells as well and could be the subject of further investigation into BCL6 and NOTCH cross-talk.

During B-cell development, induction of NOTCH2 activity by its ligand DLL1 in the murine splenic vasculature directs B cells toward marginal zone differentiation and away from the pool of naïve follicular B cells from which GCs form (40). We confirm that NOTCH2, MAML1, and several key NOTCH targets are downregulated in GC B cells in comparison to their precursor naïve B cells. Indeed, we show that NOTCH2 must be silenced for the development of fully established GCs, and that it is the transcription factor BCL6, which is a master regulator of GC formation, that mediates this downregulation. Induction of BCL6 through IL21 and IL4 in human and murine naïve B cells (23, 24) thus resulted in downregulation of NOTCH2, MAML2, and RBPJ, suggesting additional mechanisms through which NOTCH2 activity is controlled in divergent cell fates in secondary lymphoid tissues. BCL6 repression of NOTCH2 in FL is therefore derived from a normal function of BCL6 in GC B cells. It is notable that studies in human GC B cells cultured in the presence of the follicular dendritic HK cell line showed that Jg1-mediated NOTCH signaling contributes to the survival mediated by follicular dendritic cells (32). In murine B cells, NOTCH signaling through DLL1 was shown to enhance formation of IgG1 class switched plasma cells (41). However, in experiments with B cells in their physiologic context, we show that induction of ICN2 profoundly suppresses GC formation. Along with NOTCH2, BCL6 represses other growth and survival genes in GC B cells, including BCL2 and MYC (10). Collectively, the data suggest a scenario whereby BCL6 may attenuate NOTCH2 signaling in GC centroblasts in GC dark zone, but later, as BCL6 levels are downregulated and B cells undergo class switch recombination and interact with follicular dendritic cells of the GC as they transition to memory or plasma cells, the NOTCH pathway may cooperate with other survival signals to maintain the survival and proliferation of post-GC B cells. Along these lines, Lee and colleagues (42) reported five cases of activating NOTCH2 mutations in non-GCB DLBCLs, which originate from post-GC B cells. These findings underline the importance of cell context in determining whether certain genes function as oncogenes or tumor suppressors.

It should be noted that a recent study reported five NOTCH1 and two NOTCH2 gain-of-function mutations in FL, accounting for a total of 6.3% among 112 patients with FL. However, NOTCH-mutated FL cases more frequently included a DLBCL histologic component than the WT cases (43). Hence, these mutations could occur in very specific subsets of patients who may be borderline DLBCL. Along these lines, Dr. Paulli's work points to a bias toward patients with DLBCL positive for hepatitis C virus who carry NOTCH2 mutations (20%) versus patients negative for the virus (44). They could also be linked to FLs with features of late GC lymphomas. Cell-type context is clearly important because even though NOTCH activating mutations are known to drive T-cell leukemias, NOTCH is by contrast a tumor suppressor in myeloid leukemia (45). Suppression of NOTCH is evidently a critical function of BCL6 in FLs, because NOTCH2 siRNA or antagonist could rescue cell death induced by BCL6 blockade, and NOTCH2 expression killed FL cells. NOTCH2 is thus a growth suppressor of FLs, and BCL6 mediates FL pathogenesis in part through suppression of this pathway.

Finally, we show that BCL6 is a bona fide therapeutic target in FL, and not just a passenger marker. This was confirmed using two different loss-of-function strategies (siRNA and RI-BPI) and was relevant not only to cell lines but also to primary human BCL6-positive FLs. BCL6 inhibitors also induced NOTCH2 and suppressed the growth of FL xenografts, suggesting that cell-autonomous inhibition of an FL survival pathway is able to suppress FL tumor growth in vivo. This is an example of “non-oncogene” dependence, in that even though the BCL6 locus is not usually mutated in FL, cells that express it are biologically dependent on its continued presence to maintain their survival. Because RI-BPI blocks only the BTB domain lateral groove and does not affect other BCL6 functions, FL survival is clearly dependent on BCL6 recruitment of corepressors to the BCL6 BTB domain (7). The presented data suggest that BCL6 targeted therapy could be a useful approach for treatment of FLs. Importantly, because RI-BPI affects only certain BCL6 functions (46), it does not induce toxicity or inflammation in animals even when administered long term and so is suitable as a therapeutic agent for diseases that might require chronic dosing (7). Likewise, animals engineered to express a BCL6 mutant that mimics the loss of function induced by RI-BPI live normal healthy lives (47). The data expand the spectrum of patients who are candidates for RI-BPI or other BCL6 inhibitor clinical trials and offer a potential approach for more effectively eradicating these incurable tumors.

Gene Expression Microarray Data and RNA-seq

Publicly available gene expression microarray data were obtained from 191 primary FLs (9). Data processing and normalization were performed as previously described (9). For the examination of available gene expression profiles obtained from five independent sets of NB and GC B cells, data publicly available were obtained from GC B-cell array accession number GSE2350 (22). BCL6 ChIP-on-chip data have been submitted to GEO GSE29165.

Cell Lines and Reagents

DoHH2, Sc-1, WSU-DLCL2, and SU-DHL-4 cell lines were obtained from the DSMZ German collection of microorganisms and cell cultures. They were grown in RPMI-1640 medium (CellGro) supplemented with 10% FBS, 1% penicillin/streptomycin, 2 mmol/L l-glutamine and 10 mmol/L HEPES (all from Gibco). OCI-Ly1 and OCI-Ly8 cell lines obtained from OCI were grown in Iscove's Medium (CellGro) supplemented with 10% FBS, 1% penicillin/streptomycin as above. Stable cultures for DoHH2 and Sc-1 cell lines were established by retroviral infection of pMIGR1-GFP control and pMIGR1-ICN2-GFP (the intracellular domain of NOTCH2 protein that is fused to GFP). The OP9 cell line was grown in DMEM (CellGro) supplemented with 20% FBS, 1% penicillin/streptomycin, 2 mmol/L l-glutamine, 10 mmol/L HEPES, 55 μmol/L β-mercaptoethanol, and 50 μg/mL gentamicin (Gibco).

We performed DNA genotyping to identify and authenticate all the cell lines before use, December 2016 being last time they were authenticated. DNA extraction, short repeat profiling, and comparison with known cell line profiles from ATCC were performed by BioSynthesis Inc.

The RI-BPI corresponds to sequence S6.2 as previously published (7). Control and RI-BPI peptides were synthesized by Biosynthesis Inc.

Primary B-cell Populations' Isolation, Culture Conditions, and Cytokine Treatments

Tonsillar tissue was obtained as discarded material from routine tonsillectomies at the Montefiore Children's Hospital and Weill Cornell Medical College (with the approval of the Institutional Review Boards of Albert Einstein College of Medicine, Montefiore Hospital, and Weill Cornell Medical College and in accordance with the Helsinki protocols). Briefly, tonsillar mononuclear cells were separated by density centrifugation with Fico/Lite LymphoH (Atlanta Biologicals). Samples were divided in two and the mononuclear cells rophatPro Separator system as follows: Naïve B cells were stained sequentially with anti–IgD-FITC followed by FITC microbeads followed by a positive selection. GC B-cell isolation was done sequentially with anti-CD77, followed by anti-MARM, and rat anti-mouse IgG1 microbeads followed by a positive selection. The purity of the isolated B-cell populations was determined by flow-cytometry LSRII system. Naïve B cells were IgD+CD38lo and GC B cells were IgDCD77+CD38hi (see Supplementary Fig. S3 for purity of samples). The FlowJo software from Treestar, Inc. was used for the flow-cytometry analysis. Following purification, the samples were processed for mRNA and protein extraction. Naïve B cells were cocultured with a stromal layer of OP9. Cocultures were grown in RPMI with 20% FBS, 1% penicillin/streptomycin, 2 mmol/L l-glutamine and 10 mmol/L HEPES. Cytokine treatment was done using 30 ng/mL of IL4 and 30 ng/mL of IL21 or vehicle for up to 4 days. Cell viability was assessed every day by Trypan blue dye exclusion, and cytokines were added to media every other day. Alternatively, resting B lymphocytes from BL57/6 mouse spleens were isolated. B220+ splenocytes were obtained by negative selection with anti-CD43 and anti–Mac-1/CD11b monoclonal antibodies coupled to magnetic microbeads. Additional information is given in the Methods section of Supplementary Figures.

ICN2 Knock-In Mice and NP and SRBC Immunization

Experiments were performed in accordance with the guidelines of the New York University Institutional Animal Care and Use Committee. ROSA26-ICN2-IRES-YFP knock-in mice were generated by insertion of a LoxP flanked splice acceptor NEO-ATG cassette with two polyA sites followed by the ICN2-IRES-YFP cassette into the ROSA26 locus, allowing the ROSA26 promoter to drive the expression of the NEO-ATG cassette. In order to express the transgene (ICN2-IRES-YFP), these mice were crossed with tamoxifen-inducible ROSA26-CreERT2 mice expressing CreERT2 from the ubiquitously expressed ROSA26 promoter or with the CγCre. For GC studies, serum of preimmunized mice (ROSA 26 WT and ROSA26-ICN2-IRES-YFP knock-in) was collected 1 day prior to the start of the experiment (day −1). The next day, mice were injected intraperitoneally with 100 μL of 4Hydroxy-3nitrophenylacetyl hapten-chicken gamma globulin (NP65-GCC) in alum (day 0). For CreERT2 induction, tamoxifen was solubilized in corn oil at a concentration of 20 mg/mL, and 1 day after NP immunization, a single intraperitoneal injection of 0.2 mg/g body weight was administered. Alternatively, mice were immunized intraperitoneally with 500 μL 2% SRBC in PBS. Serum was collected at days 7 and 14. Mice were sacrificed, and spleens were harvested for IHC and flow-cytometry analysis. A schematic representation of the experiment is given in Supplementary Fig. S4A. Genotyping primers are listed in Supplementary Table S5. Additional information is available in the Methods section of Supplementary Figures.

Flow Cytometry and Immunohistochemistry of GC

B220+ splenocytes from WT and ICN2 mice were obtained as described above. For the GC B-cell population, we gated on B220+ and GL7 eFluor674 and CD95(Fas) PECy7. For plasma cell population (CD38+ CD138+), we used CD38 APC and CD138 PE. ICN2 expression was assessed by B220+ YFP+. Immunohistochemistry of spleens was performed on formalin-fixed paraffin-embedded sections with the following primary antibodies: PNA, BCL6 (N3), and CD45R/B220. BCL6 shows a purple pattern, whereas B220 staining is pale brown. To count the GC, we used CellSens Software (Olympus America Inc.). Additional information is available in the Methods section of Supplementary Figures.

Primary Lymphoma Samples

Deidentified primary FL specimen tissues were obtained in accordance with the guidelines and approval of the Institutional Review Board of Weill Cornell Medical College and in accordance with the Helsinki protocols. We selected the specimens based on estimated tumor content >80% by our collaborating pathologist Dr. Wayne Tam. Single-cell suspensions from lymph node biopsies were obtained by physical disruption of tissues (using scalpels and cell strainers), followed by cell density gradient separation (Fico/Lite LymphoH; Atlanta Biologicals). Cell number and viability were determined by Trypan blue dye exclusion, and cells were cultivated in medium containing 80% RPMI and 20% human serum supplemented with antibiotics, l-glutamine 4 mmol/L and HEPES 10 mmol/L. Cells were exposed in duplicate to control and RI-BPI at indicated concentrations for 48 hours. Viability was determined as detailed above. The BCL6 protein status was determined in paraffin-embedded samples by immunohistochemistry using anti-BCL6 (Dako North America).

Mice Xenotransplant Studies

All animal procedures followed NIH protocols and were approved by the Animal Institute Committee of the Weill Cornell Medical College. Six- to 8-week-old male SCID mice were purchased from the National Cancer Institute (NCI) and housed in a clean environment. Mice were subcutaneously injected in the left flank with low-passage 107 human lymphoma cells (DoHH2 and Sc-1). Tumor volume was monitored every other day using electronic digital calipers in two dimensions. Tumor volume was calculated using the formula: Tumor volume (mm3) = (smallest diameter2 × largest diameter)/2. When tumors reached a palpable size (approximately 75–100 mm3 after 21 days post-injection), the mice were randomized to two different treatment arms. RI-BPI was stored lyophilized at −20°C until reconstituted with sterile pure water immediately before use. RI-BPI was administered by intraperitoneal injection. Mice were weighed twice a week. All mice were euthanized by cervical dislocation under anesthesia when at least 2 of 10 tumors reached 20 mm in any dimension (equivalent to 1 g), which was generally on day 9 or 10 of the treatment schedule. At the moment of euthanasia, the tumors and other tissues were harvested and weighed.

Transfections, Anti-NRR2, and RI-BPI Treatment of Lymphoma Cell Lines

For siRNA knockdown experiments, BCL6-specific siRNAs (Cat# HSS100966) and control nontargeting siRNA (Cat# 1299003) were purchased from Invitrogen. siRNA sequences for NOTCH2 (Cat# J-012235) were purchased from Dharmacon-ThermoScientific. siRNA (20 pmol/L) was suspended in 20 μL of Solution SF and introduced into 3 × 106 cells using the Amaxa 96-well nucleofector (Lonza). For Western blot experiments, rabbit antibody raised against BCL6-N3 (sc-858) and anti–Actin-HRP conjugated (sc-1615) were purchased from Santa Cruz Biotechnology; rabbit antibody raised against ICN2 (ab72803) that recognizes the cleaved intracellular fragment of NOTCH2 was purchased from Abcam. For experiments using cell lines treated with RI-BPI, 10 to 20 μmol/L final concentration of the drug was used for 24 and 48 hours. Doses were selected based on the relative GI50s for each cell line. For DoHH2 and Sc-1 anti-NRR2 experiments, 2 × 106 cells were treated with 2 μg/mL of anti-NRR2 from Genentech, Inc. or a negative control, anti-IgG1 from Southern Biotech. Twenty-four hours after treatment, cells were replated to 96-well plates and RI-BPI was added. Cell viability was measured 24 or 48 hours after RI-BPI treatment as detailed below. Experiments were performed in triplicate, and the figures represent the average of three experiments ± SEM.

Cell Viability Assay and Growth Inhibition Determination

Cell viability on lymphoma cell lines was determined using a fluorometric resazurin reduction method (CellTiter-Blue, Promega) and relative fluorescence (560exitation/590emission) detected with a Synergy4 Microplate Reader (BioTek Instruments). The number of viable cells was calculated by extrapolating from the standard curve. Fluorescence was measured for three replicates per treatment condition and cell viability in drug-treated cells normalized to their respective controls. Experiments were performed in triplicate. The figures represent the average of three experiments and the SEM. To determine growth inhibition of lymphoma cell lines exposed to different doses of RI-BPI, cells were plated at concentrations sufficient to keep untreated cells in exponential growth over the 48-hour drug exposure time. Cell viability was measured as described above. Trypan blue dye exclusion was used as a secondary method to confirm the results. Fluorescence was determined for 6 replicates per treatment condition, and cell viability in drug-treated cells was normalized to their respective controls. Unless stated otherwise, the experiments were carried out in biological triplicate. The CompuSyn software package (Biosoft) was used to plot dose–effect curves and determine the drug concentration that inhibits the growth of cell lines by 50% compared with control (GI50). The linear correlation coefficient was higher than 0.90 for each curve in the median-effect plot.

Additional experimental procedures including quantitative RT-PCR, plasmids and reporter assays, ChIP and ChIP-on-chip assay, bioinformatics analysis of Gene Expression data, primary cultures, flow cytometry, and mouse studies can be found on Supplemental Experimental Procedures.

A. Melnick reports receiving commercial research grants from Janssen, GSK, Lilly, and Roche, and is a consultant/advisory board member for Epizyme and Roche. No potential conflicts of interest were disclosed by the other authors.

Conception and design: E. Valls, H. Geng, L. Cerchietti, I. Aifantis, A. Melnick

Development of methodology: E. Valls, H. Geng, M. Rivas, S.N. Yang, X. Agirre, W. Ci, A. Melnick

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): E. Valls, C. Lobry, L. Cerchietti, P. Oh, S.N. Yang, E. Oswald, C.W. Graham, Y. Jiang, K. Hatzi, E. Perkey, Z. Li, W. Tam, K. Bhatt, P.A. Zweidler-McKay, I. Maillard, W. Ci, A. Melnick

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): E. Valls, C. Lobry, H. Geng, M. Cardenas, L. Cerchietti, E. Oswald, Y. Jiang, K. Hatzi, X. Agirre, Z. Li, I. Maillard, O. Elemento, W. Ci, I. Aifantis, A. Melnick

Writing, review, and/or revision of the manuscript: E. Valls, C. Lobry, P. Oh, W. Tam, J.P. Leonard, I. Aifantis, A. Melnick

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): E. Valls, L. Wang, M. Cardenas, W. Tam, K. Bhatt, J.P. Leonard, I. Aifantis

Study supervision: L. Cerchietti, A. Melnick

We thank the Weill Cornell Epigenomics Core Facility for support with ChIP-on-chip assays. We thank Dr. Artavanis-Tsakonas for sharing the ROSA26-NOTCH2IC animals.

This research was funded by a follicular lymphoma research grant from the Lymphoma Research Foundation to A. Melnick. A. Melnick is also supported by NCIR01CA104348, NCIR01CA143032, and the Chemotherapy Foundation. E. Valls has been the recipient of an IDIBAPS Postdoctoral Fellowship-BIOTRACK, supported by the European Community's Seventh Framework Programme (EC FP7/2007-2013) under the grant agreement number 229673. I. Aifantis is supported by the NIH (RO1CA216421, RO1CA169784, RO1CA202025, R01CA133379, and R01CA149655) and the NYSTEM program of the New York State Health Department. C. Lobry is supported by a Leukemia and Lymphoma Society Career Development Award and a French government ATIP-Avenir grant. W. Ci is supported by the National Basic Research Programme (Grant No. 2016YFC0900303); the National Natural Science Foundation of China (Grant No. 81422035, 91519307, and 81672541); and the Chinese Academy of Science (CAS; Grant No. QYZDB-SSW-SMC039 and KJZD-EW-L14).

1.
Tan
D
,
Horning
SJ
. 
Follicular lymphoma: clinical features and treatment
.
Hematol Oncol Clin North Am
2008
;
22
:
863
82
,
viii
.
2.
Piccaluga
PP
,
Sapienza
MR
,
Agostinelli
C
,
Sagramoso
C
,
Mannu
C
,
Sabattini
E
, et al
Biology and treatment of follicular lymphoma
.
Expert Rev Hematol
2009
;
2
:
533
47
.
3.
Egle
A
,
Harris
AW
,
Bath
ML
,
O'Reilly
L
,
Cory
S
. 
VavP-Bcl2 transgenic mice develop follicular lymphoma preceded by germinal center hyperplasia
.
Blood
2004
;
103
:
2276
83
.
4.
Skinnider
BF
,
Horsman
DE
,
Dupuis
B
,
Gascoyne
RD
. 
Bcl-6 and Bcl-2 protein expression in diffuse large B-cell lymphoma and follicular lymphoma: correlation with 3q27 and 18q21 chromosomal abnormalities
.
Hum Pathol
1999
;
30
:
803
8
.
5.
Swerdlow
SH
,
Campo
E
,
Harris
NL
,
Jaffe
ES
,
Pileri
SA
,
Thiele
J
, et al
WHO classification of tumours of haematopoietic and lymphoid tissues
, 4th ed.
Geneva, Switzerland
:
WHO Press
; 
2008
.
6.
Hatzi
K
,
Melnick
A
. 
Breaking bad in the germinal center: how deregulation of BCL6 contributes to lymphomagenesis
.
Trends Mol Med
2014
;
20
:
343
52
.
7.
Cerchietti
LC
,
Yang
SN
,
Shaknovich
R
,
Hatzi
K
,
Polo
JM
,
Chadburn
A
, et al
A peptomimetic inhibitor of BCL6 with potent antilymphoma effects in vitro and in vivo
.
Blood
2009
;
113
:
3397
405
.
8.
Polo
JM
,
Dell'Oso
T
,
Ranuncolo
SM
,
Cerchietti
L
,
Beck
D
,
Da Silva
GF
, et al
Specific peptide interference reveals BCL6 transcriptional and oncogenic mechanisms in B-cell lymphoma cells
.
Nat Med
2004
;
10
:
1329
35
.
9.
Polo
JM
,
Juszczynski
P
,
Monti
S
,
Cerchietti
L
,
Ye
K
,
Greally
JM
, et al
Transcriptional signature with differential expression of BCL6 target genes accurately identifies BCL6-dependent diffuse large B cell lymphomas
.
Proc Natl Acad Sci U S A
2007
;
104
:
3207
12
.
10.
Ci
W
,
Polo
JM
,
Cerchietti
L
,
Shaknovich
R
,
Wang
L
,
Yang
SN
, et al
The BCL6 transcriptional program features repression of multiple oncogenes in primary B cells and is deregulated in DLBCL
.
Blood
2009
;
113
:
5536
48
.
11.
Elemento
O
,
Slonim
N
,
Tavazoie
S
. 
A universal framework for regulatory element discovery across all genomes and data types
.
Mol Cell
2007
;
28
:
337
50
.
12.
Shaffer
AL
,
Yu
X
,
He
Y
,
Boldrick
J
,
Chan
EP
,
Staudt
LM
. 
BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control
.
Immunity
2000
;
13
:
199
212
.
13.
Alizadeh
AA
,
Eisen
MB
,
Davis
RE
,
Ma
C
,
Lossos
IS
,
Rosenwald
A
, et al
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling
.
Nature
2000
;
403
:
503
11
.
14.
Whitfield
ML
,
Sherlock
G
,
Saldanha
AJ
,
Murray
JI
,
Ball
CA
,
Alexander
KE
, et al
Identification of genes periodically expressed in the human cell cycle and their expression in tumors
.
Mol Biol Cell
2002
;
13
:
1977
2000
.
15.
Palomero
T
,
Lim
WK
,
Odom
DT
,
Sulis
ML
,
Real
PJ
,
Margolin
A
, et al
NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth
.
Proc Natl Acad Sci U S A
2006
;
103
:
18261
6
.
16.
Shaffer
AL
,
Lin
KI
,
Kuo
TC
,
Yu
X
,
Hurt
EM
,
Rosenwald
A
, et al
Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program
.
Immunity
2002
;
17
:
51
62
.
17.
Dave
SS
,
Wright
G
,
Tan
B
,
Rosenwald
A
,
Gascoyne
RD
,
Chan
WC
, et al
Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells
.
N Engl J Med
2004
;
351
:
2159
69
.
18.
Sharma
VM
,
Calvo
JA
,
Draheim
KM
,
Cunningham
LA
,
Hermance
N
,
Beverly
L
, et al
Notch1 contributes to mouse T-cell leukemia by directly inducing the expression of c-myc
.
Mol Cell Biol
2006
;
26
:
8022
31
.
19.
Weng
AP
,
Millholland
JM
,
Yashiro-Ohtani
Y
,
Arcangeli
ML
,
Lau
A
,
Wai
C
, et al
c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma
.
Genes Dev
2006
;
20
:
2096
109
.
20.
Dave
SS
,
Fu
K
,
Wright
GW
,
Lam
LT
,
Kluin
P
,
Boerma
EJ
, et al
Molecular diagnosis of Burkitt's lymphoma
.
N Engl J Med
2006
;
354
:
2431
42
.
21.
Saito
T
,
Chiba
S
,
Ichikawa
M
,
Kunisato
A
,
Asai
T
,
Shimizu
K
, et al
Notch2 is preferentially expressed in mature B cells and indispensable for marginal zone B lineage development
.
Immunity
2003
;
18
:
675
85
.
22.
Basso
K
,
Margolin
AA
,
Stolovitzky
G
,
Klein
U
,
Dalla-Favera
R
,
Califano
A
. 
Reverse engineering of regulatory networks in human B cells
.
Nat Genet
2005
;
37
:
382
90
.
23.
Linterman
MA
,
Beaton
L
,
Yu
D
,
Ramiscal
RR
,
Srivastava
M
,
Hogan
JJ
, et al
IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses
.
J Exp Med
2010
;
207
:
353
63
.
24.
Tsuruoka
N
,
Arima
M
,
Arguni
E
,
Saito
T
,
Kitayama
D
,
Sakamoto
A
, et al
Bcl6 is required for the IL-4-mediated rescue of the B cells from apoptosis induced by IL-21
.
Immunol Lett
2007
;
110
:
145
51
.
25.
Oh
P
,
Lobry
C
,
Gao
J
,
Tikhonova
A
,
Loizou
E
,
Manent
J
, et al
In vivo mapping of notch pathway activity in normal and stress hematopoiesis
.
Cell Stem Cell
2013
;
13
:
190
204
.
26.
Casola
S
,
Cattoretti
G
,
Uyttersprot
N
,
Koralov
SB
,
Seagal
J
,
Hao
Z
, et al
Tracking germinal center B cells expressing germ-line immunoglobulin gamma1 transcripts by conditional gene targeting
.
Proc Natl Acad Sci U S A
2006
;
103
:
7396
401
.
27.
Hatzi
K
,
Jiang
Y
,
Huang
C
,
Garrett-Bakelman
F
,
Gearhart
MD
,
Giannopoulou
EG
, et al
A hybrid mechanism of action for BCL6 in B cells defined by formation of functionally distinct complexes at enhancers and promoters
.
Cell Rep
2013
;
4
:
578
88
.
28.
Cerchietti
LC
,
Hatzi
K
,
Caldas-Lopes
E
,
Yang
SN
,
Figueroa
ME
,
Morin
RD
, et al
BCL6 repression of EP300 in human diffuse large B cell lymphoma cells provides a basis for rational combinatorial therapy
.
J Clin Invest
2010
;
120
:
4569
82
.
29.
Cerchietti
LC
,
Polo
JM
,
Da Silva
GF
,
Farinha
P
,
Shaknovich
R
,
Gascoyne
RD
, et al
Sequential transcription factor targeting for diffuse large B-cell lymphomas
.
Cancer Res
2008
;
68
:
3361
9
.
30.
Ortega-Molina
A
,
Boss
IW
,
Canela
A
,
Pan
H
,
Jiang
Y
,
Zhao
C
, et al
The histone lysine methyltransferase KMT2D sustains a gene expression program that represses B cell lymphoma development
.
Nat Med
2015
;
21
:
1199
208
.
31.
Jiang
Y
,
Ortega-Molina
A
,
Geng
H
,
Ying
HY
,
Hatzi
K
,
Parsa
S
, et al
CREBBP inactivation promotes the development of HDAC3-dependent lymphomas
.
Cancer Discov
2017
;
7
:
38
53
.
32.
Yoon
SO
,
Zhang
X
,
Berner
P
,
Blom
B
,
Choi
YS
. 
Notch ligands expressed by follicular dendritic cells protect germinal center B cells from apoptosis
.
J Immunol
2009
;
183
:
352
8
.
33.
Tran
IT
,
Sandy
AR
,
Carulli
AJ
,
Ebens
C
,
Chung
J
,
Shan
GT
, et al
Blockade of individual Notch ligands and receptors controls graft-versus-host disease
.
J Clin Invest
2013
;
123
:
1590
604
.
34.
Wu
Y
,
Cain-Hom
C
,
Choy
L
,
Hagenbeek
TJ
,
de Leon
GP
,
Chen
Y
, et al
Therapeutic antibody targeting of individual Notch receptors
.
Nature
2010
;
464
:
1052
7
.
35.
Lee
CH
,
Chawla
A
,
Urbiztondo
N
,
Liao
D
,
Boisvert
WA
,
Evans
RM
, et al
Transcriptional repression of atherogenic inflammation: modulation by PPARdelta
.
Science
2003
;
302
:
453
7
.
36.
Vasanwala
FH
,
Kusam
S
,
Toney
LM
,
Dent
AL
. 
Repression of AP-1 function: a mechanism for the regulation of Blimp-1 expression and B lymphocyte differentiation by the B cell lymphoma-6 protooncogene
.
J Immunol
2002
;
169
:
1922
9
.
37.
Zweidler-McKay
PA
,
He
Y
,
Xu
L
,
Rodriguez
CG
,
Karnell
FG
,
Carpenter
AC
, et al
Notch signaling is a potent inducer of growth arrest and apoptosis in a wide range of B-cell malignancies
.
Blood
2005
;
106
:
3898
906
.
38.
Sakano
D
,
Kato
A
,
Parikh
N
,
McKnight
K
,
Terry
D
,
Stefanovic
B
, et al
BCL6 canalizes Notch-dependent transcription, excluding Mastermind-like1 from selected target genes during left-right patterning
.
Dev Cell
2010
;
18
:
450
62
.
39.
Tiberi
L
,
van den Ameele
J
,
Dimidschstein
J
,
Piccirilli
J
,
Gall
D
,
Herpoel
A
, et al
BCL6 controls neurogenesis through Sirt1-dependent epigenetic repression of selective Notch targets
.
Nat Neurosci
2012
;
15
:
1627
35
.
40.
Pillai
S
,
Cariappa
A
. 
The follicular versus marginal zone B lymphocyte cell fate decision
.
Nat Rev Immunol
2009
;
9
:
767
77
.
41.
Thomas
M
,
Calamito
M
,
Srivastava
B
,
Maillard
I
,
Pear
WS
,
Allman
D
. 
Notch activity synergizes with B-cell-receptor and CD40 signaling to enhance B-cell activation
.
Blood
2007
;
109
:
3342
50
.
42.
Lee
SY
,
Kumano
K
,
Nakazaki
K
,
Sanada
M
,
Matsumoto
A
,
Yamamoto
G
, et al
Gain-of-function mutations and copy number increases of Notch2 in diffuse large B-cell lymphoma
.
Cancer Sci
2009
;
100
:
920
6
.
43.
Karube
K
,
Martinez
D
,
Royo
C
,
Navarro
A
,
Pinyol
M
,
Cazorla
M
, et al
Recurrent mutations of NOTCH genes in follicular lymphoma identify a distinctive subset of tumours
.
J Pathol
2014
;
234
:
423
30
.
44.
Arcaini
L
,
Rossi
D
,
Lucioni
M
,
Nicola
M
,
Bruscaggin
A
,
Fiaccadori
V
, et al
The NOTCH pathway is recurrently mutated in diffuse large B cell lymphoma associated with hepatitis C virus infection
.
Haematologica
2014
;
100
:
246
52
.
45.
Klinakis
A
,
Lobry
C
,
Abdel-Wahab
O
,
Oh
P
,
Haeno
H
,
Buonamici
S
, et al
A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia
.
Nature
2011
;
473
:
230
3
.
46.
Parekh
S
,
Polo
JM
,
Shaknovich
R
,
Juszczynski
P
,
Lev
P
,
Ranuncolo
SM
, et al
BCL6 programs lymphoma cells for survival and differentiation through distinct biochemical mechanisms
.
Blood
2007
;
110
:
2067
74
.
47.
Huang
C
,
Hatzi
K
,
Melnick
A
. 
Lineage-specific functions of BCL6 in immunity and inflammation are mediated through distinct biochemical mechanisms
.
Nat Immunol
2013
;
14
:
380
8
.

Supplementary data