scholarly journals Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells

Blood ◽  
2010 ◽  
Vol 115 (5) ◽  
pp. 975-984 ◽  
Author(s):  
Katia Basso ◽  
Masumichi Saito ◽  
Pavel Sumazin ◽  
Adam A. Margolin ◽  
Kai Wang ◽  
...  

Abstract BCL6 is a transcriptional repressor required for mature B-cell germinal center (GC) formation and implicated in lymphomagenesis. BCL6's physiologic function is only partially known because the complete set of its targets in GC B cells has not been identified. To address this issue, we used an integrated biochemical-computational-functional approach to identify BCL6 direct targets in normal GC B cells. This approach includes (1) identification of BCL6-bound promoters by genome-wide chromatin immunoprecipitation, (2) inference of transcriptional relationships by the use of a regulatory network reverse engineering approach (ARACNe), and (3) validation of physiologic relevance of the candidate targets down-regulated in GC B cells. Our approach demonstrated that a large set of promoters (> 4000) is physically bound by BCL6 but that only a fraction of them is repressed in GC B cells. This set of 1207 targets identifies several cellular functions directly controlled by BCL6 during GC development, including activation, survival, DNA-damage response, cell cycle arrest, cytokine signaling, Toll-like receptor signaling, and differentiation. These results define a broad role of BCL6 in preventing centroblasts from responding to signals leading to exit from the GC before they complete the phase of proliferative expansion and of antibody affinity maturaton.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 212-212 ◽  
Author(s):  
Ryan T. Phan ◽  
Huifeng Niu ◽  
Masumichi Saito ◽  
Katia Basso ◽  
Giorgio Cattoretti ◽  
...  

Abstract The proto-oncogene BCL6 encodes a BTB/POZ-zinc finger transcriptional repressor that is necessary for germinal center (GC) formation and is implicated in the pathogenesis of B-cell lymphoma. In ~50% diffuse large cell lymphoma and 10% follicular lymphoma, BCL6 gene expression is deregulated by chromosomal translocations or mutations that affect its 5′ regulatory region. The precise function of BCL6 in GC development and lymphomagenesis is unclear since very few BCL6 direct target genes have been identified. We report that BCL6 suppresses p53-dependent and p53-indepenent growth arrest and apoptosis responses in GC B cells. BCL6 directly suppresses the transcription of the p53 gene, as demonstrated by (1) chromatin immunoprecipitation (ChIP) assays showing that BCL6 binds the p53 promoter region in vivo; and (2) transient transfection/reporter assays identifying within the p53 promoter region two BCL6-binding sites that mediate BCL6-mediated suppression of p53 transcription. Accordingly, suppression of BCL6 expression via specific siRNA leads to increased expression of p53 both under basal condition and in response to DNA damage. Consistent with a physiological role for BCL6-mediated p53 suppression, immunohistochemical analysis shows that p53 expression is absent in GC B cells where BCL6 is highly expressed. In addition, our data reveal that BCL6 inhibits the p53-independent activation of the p21/WAF1 cell cycle arrest gene by binding to Miz-1, a transcription factor involved in p21 activation. Consistent with a role of BCL6 in inhibiting p53-related cell cycle arrest and apoptotic responses, constitutive expression of BCL6 suppresses p53 expression and p53-target genes (P21 and PUMA) and protects B cell lines from apoptosis induced by DNA damage. These results indicate that one function of BCL6 is to allow GC B cells (centroblasts) to constitutively proliferate and to sustain the physiologic DNA breaks required for immunoglobulin switch recombination and somatic hypermutation without inducing p53-related responses. These findings also imply that B cell lymphoma with deregulated BCL6 expression are functionally p53-negative and impaired in apoptotic responses.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3465-3465
Author(s):  
Irina Velichutina ◽  
Rita Shaknovich ◽  
Huimin Geng ◽  
Ari Melnick ◽  
Olivier Elemento

Abstract Abstract 3465 Poster Board III-353 EZH2 is the catalytic subunit of the PRC2 Polycomb complex and mediates transcriptional repression through its histone methyltransferase activity. It is over-expressed in many types of aggressive tumors, e.g., prostate and breast cancer and this over-expression is generally associated with poor patient prognosis. It is also over-expressed in certain lymphomas, e.g., follicular lymphoma; however its exact role and importance in normal and malignant B-cells remains unclear. Most B-cell lymphomas arise from germinal center (GC) B-cells. We thus first set out to investigate the function and activity of EZH2 in normal B-cells. We confirmed a previously published observation that expression of EZH2 protein is greatly elevated during developmental transition from resting Naive B-cells to rapidly proliferating GC B-lymphocytes. Using ChIP-on-chip, we then determined the genome-wide pattern of EZH2 binding in GC B cells and found that EZH2 targets ∼1,800 promoters in these cells (at FDR<0.1). We also mapped the genome-wide distribution of H3K27me3 histone marks in GC B cells; consistent with the known role of EZH2 in catalyzing H3K27me3, a majority (62%) of EZH2 target promoters also displayed a very strong H3K27me3 peak (p=0, hypergeometric test). Also consistent with the repressive nature of H3K27me3, expression arrays rezvealed that EZH2 targets are usually expressed at a lower level in GC B cells than other genes (p<1e-80). However, contrary to a previously postulated role of EZH2 in promoting DNA methylation, our results indicate that EZH2-bound promoters are largely hypomethylated in GC B cells (p=0, Wilcoxon test). From the biological standpoint EZH2 target genes are enriched with transcription factors (p<1e-5), kinases (p<1e-6), and other components of signal transduction pathways such as TGF-beta, WNT, EGFR, PDGFR, and VEGF. EZH2 also targets and represses many tumor suppressor genes, e.g., CDKN1A/p21 and CDKN1B/p27, CDKN2A/p16 and CDKN2A/p14. Using an unbiased motif discovery procedure, we associate EZH2 binding with sequences highly similar to those bound by orthologous PRC2 in Drosophila; we find that EZH2 binding is also associated with the highly statistically significant depletion of regulatory sequences typically bound by transcriptional activators. We then compared the genome-wide binding patterns of EZH2 in GC B cells and embryonic stem cells and observed a strong overlap of EZH2 targets between these cell types (>30% of GC B cells targets are also bound in hESCs, p<1e-378). However, we also observed a large GC B cell-specific EZH2 regulatory program with >1,000 genes. Seeking to extrapolate our binding data to GC-derived Diffuse Large B Cell Lymphoma (DLBCL), we found that the expression profile of many EZH2 target genes is anti-correlated with EZH2 mRNA levels in expression profiles of primary DLBCL tumors. Surprisingly, we found that this anti-correlation was most pronounced among GC B cell-specific EZH2 targets (p<1e-26). In turn, the EZH2 mRNA level was itself positively correlated with cellular proliferation in primary DLBCL tumors, as measured by Ki67 staining (Pearson correlation = 0.3, p<0.001). Finally siRNA-mediated down-regulation of EZH2 in SUDHL4 DLBCL cells resulted in acute cell cycle arrest at the G1/S transition in SUDHL4 cells and upregulation of EZH2 target genes with cell cycle inhibitory functions such as those mentioned above. Altogether, these data suggest a scenario whereby EZH2 upregulation in GC B-cells leads to its recruitment to genes containing Polycomb Response Elements with consequent H3K27 trimethylation and silencing of a GC B cell context specific cohort of genes including those involved in restraining cellular proliferation, thus contributing to the ability of these cells to undergo massive clonal expansion. This function of EZH2 may also contribute to the malignant transformation of GC B-cells into DLBCLs and facilitate their proliferative phenotype. Thus, our results indicate that therapeutic targeting of EZH2 might have significant anti-lymphoma effects and support the rationale for development of inhibitors of the EZH2 SET domain. Disclosures No relevant conflicts of interest to declare.


2005 ◽  
Vol 203 (1) ◽  
pp. 63-72 ◽  
Author(s):  
Chang Hoon Lee ◽  
Mark Melchers ◽  
Hongsheng Wang ◽  
Ted A. Torrey ◽  
Rebecca Slota ◽  
...  

Interferon (IFN) consensus sequence-binding protein/IFN regulatory factor 8 (IRF8) is a transcription factor that regulates the differentiation and function of macrophages, granulocytes, and dendritic cells through activation or repression of target genes. Although IRF8 is also expressed in lymphocytes, its roles in B cell and T cell maturation or function are ill defined, and few transcriptional targets are known. Gene expression profiling of human tonsillar B cells and mouse B cell lymphomas showed that IRF8 transcripts were expressed at highest levels in centroblasts, either from secondary lymphoid tissue or transformed cells. In addition, staining for IRF8 was most intense in tonsillar germinal center (GC) dark-zone centroblasts. To discover B cell genes regulated by IRF8, we transfected purified primary tonsillar B cells with enhanced green fluorescent protein–tagged IRF8, generated small interfering RNA knockdowns of IRF8 expression in a mouse B cell lymphoma cell line, and examined the effects of a null mutation of IRF8 on B cells. Each approach identified activation-induced cytidine deaminase (AICDA) and BCL6 as targets of transcriptional activation. Chromatin immunoprecipitation studies demonstrated in vivo occupancy of 5′ sequences of both genes by IRF8 protein. These results suggest previously unappreciated roles for IRF8 in the transcriptional regulation of B cell GC reactions that include direct regulation of AICDA and BCL6.


2020 ◽  
Vol 217 (7) ◽  
Author(s):  
Amparo Toboso-Navasa ◽  
Arief Gunawan ◽  
Giulia Morlino ◽  
Rinako Nakagawa ◽  
Andrea Taddei ◽  
...  

Memory B cells (MBCs) are key for protection from reinfection. However, it is mechanistically unclear how germinal center (GC) B cells differentiate into MBCs. MYC is transiently induced in cells fated for GC expansion and plasma cell (PC) formation, so-called positively selected GC B cells. We found that these cells coexpressed MYC and MIZ1 (MYC-interacting zinc-finger protein 1 [ZBTB17]). MYC and MIZ1 are transcriptional activators; however, they form a transcriptional repressor complex that represses MIZ1 target genes. Mice lacking MYC–MIZ1 complexes displayed impaired cell cycle entry of positively selected GC B cells and reduced GC B cell expansion and PC formation. Notably, absence of MYC–MIZ1 complexes in positively selected GC B cells led to a gene expression profile alike that of MBCs and increased MBC differentiation. Thus, at the GC positive selection stage, MYC–MIZ1 complexes are required for effective GC expansion and PC formation and to restrict MBC differentiation. We propose that MYC and MIZ1 form a module that regulates GC B cell fate.


2017 ◽  
Vol 114 (18) ◽  
pp. 4751-4756 ◽  
Author(s):  
Takeharu Minamitani ◽  
Yijie Ma ◽  
Hufeng Zhou ◽  
Hiroshi Kida ◽  
Chao-Yuan Tsai ◽  
...  

Epstein–Barr virus (EBV) is a major cause of immunosuppression-related B-cell lymphomas and Hodgkin lymphoma (HL). In these malignancies, EBV latent membrane protein 1 (LMP1) and LMP2A provide infected B cells with surrogate CD40 and B-cell receptor growth and survival signals. To gain insights into their synergistic in vivo roles in germinal center (GC) B cells, from which most EBV-driven lymphomas arise, we generated a mouse model with conditional GC B-cell LMP1 and LMP2A coexpression. LMP1 and LMP2A had limited effects in immunocompetent mice. However, upon T- and NK-cell depletion, LMP1/2A caused massive plasmablast outgrowth, organ damage, and death. RNA-sequencing analyses identified EBV oncoprotein effects on GC B-cell target genes, including up-regulation of multiple proinflammatory chemokines and master regulators of plasma cell differentiation. LMP1/2A coexpression also up-regulated key HL markers, including CD30 and mixed hematopoietic lineage markers. Collectively, our results highlight synergistic EBV membrane oncoprotein effects on GC B cells and provide a model for studies of their roles in immunosuppression-related lymphoproliferative diseases.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 932-932
Author(s):  
Jeff X. Zhou ◽  
Chang-Hoon Lee ◽  
Chen-Feng Qi ◽  
Zohreh Maghashfar ◽  
Ming Zhao ◽  
...  

Abstract The transcription factor, interferon regulatory factor 8 (IRF8), is expressed at low levels in both naïve and terminally differentiated B cells but at high levels in germinal center (GC) B cells where it contributes to transcriptional activation of two genes critically involved in the GC reaction: BCL6, a BTB/POZ-zinc finger transcriptional repressor; and AICDA (also known as AID) a single stranded DNA deaminase (Lee et al. J. Exp. Med.201(1):63–72, 2006). AID is responsible for the generation of double stranded DNA breaks (DSDB) that occur physiologically in B cells during the processes of somatic hypermutation (SHM) of immunoglobulin (Ig) gene variable region sequences and Ig class switch recombination (CSR). The response of most cell types to DSDB is marked by activation of p53 (also termed TP53) that induces cell cycle arrest or apoptosis. p53 also functions at a critical checkpoint to prevent aberrant repair of DSDB in Ig genes that can result in chromosomal translocations that activate proto-oncogenes. GC B cells, however, must be able to tolerate these breaks without experiencing p53-dependent apoptosis. Recent studies showed that functional inactivation of p53 in GC cells is governed in part by transcriptional repression of the p53 gene by BCL6 (Phan and Dalla-Favera Nature432(7017):635–639, 2004). Here we report that IRF8 also suppresses p53 function via transcriptional activation of MDM2. MDM2 is a p53-binding protein and E3 ubiquitin ligase that blocks the transcriptional activity of p53 and stimulates p53 degradation. The levels of both Mdm2 mRNA and its encoded protein were decreased in GC B cells of Irf8 knockout mice as well as in a B lymphoma cell line with siRNA-induced knockdowns of IRF8 expression. Conversely, MDM2 protein levels were increased in cells induced to overexpress IRF8. Transfection of a B lymphoma cell line with an IRF8 expression vector resulted in marked activation of a Mdm2 promoter reporter construct. Oligonucleotide pull-down assays using sequences from the Mdm2 promoter similar to the interferon stimulated response element, a known IRF8 target sequence, were found to bring down IRF8. IL-21, produced by GC T helper cells, promotes the differentiation of B cells activated through the BCR, while inducing apoptosis of unengaged cells. Studies of a B lymphoma cell line treated with IL21 showed that the rate of apoptosis was significantly increased when IRF8 expression was suppressed by siRNAs. The same pattern was true for cells treated with etoposide, a drug that induces DSDB. These results indicate that IRF8 shepherds B cells through the GC reaction by stimulating expression of BCL6 and MDM2 thereby suppressing p53-mediated cell cycle arrest or death in response to DNA breaks.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 705-705
Author(s):  
Jenny Zhang ◽  
Dereje D. Jima ◽  
Cassandra L. Jacobs ◽  
Eva Gottwein ◽  
Grace Huang ◽  
...  

Abstract Background: Mature B cell differentiation provides an important mechanism for the acquisition of adaptive immunity. Malignancies derived from mature B cells are common and constitute the majority of leukemias and lymphomas. MicroRNAs are known to play a role in oncogenesis, lineage-selection, and immune cell function, including early B cell differentiation. However, the full extent and function of microRNA expression during mature B cell differentiation and in B cell malignancies are not known. Methods: From normal young patients undergoing tonsillectomies, we sorted the mature B cell subsets (naive, germinal center, memory and plasma) using FACS, based on their expression of CD19, CD38, IgD and CD27. These sorted B cells were profiled for microRNA expression using a highly sensitive multiplexed real-time PCR assay, as well as for gene expression at the whole genome level using Affymetrix U133plus microarrays. miRNA targets can be predicted based on seed sequence matching of their 2–8 nt to the 3′UTR of gene transcripts. For each B cell stage, we experimentally validated microRNA regulation of predicted target genes of interest, LMO2, MYBL1 and PRDM1, by microRNA over-expression experiments and luciferase assays. Results: We found that microRNAs have a characteristic expression pattern that defines each mature B cell stage. Examination of both microRNA and mRNA expression showed that in each B cell population, the target genes predicted based on seed matching were expressed at lower levels, results that were highly significant (P<1E-10). We found that differential microRNA expression is important at every B cell stage transition, and differentially expressed microRNAs frequently target differentially expressed transcription factors. In the naive to germinal center B cell and germinal center B cell to memory cell transitions, we found that miR-223 had an inverse relationship with its predicted target genes LMO2 and MYBL1. To test this relationship predicted based on seed pairing, in Germinal Center-derived BJAB cells, we over-expressed miR-223 by introducing its precursor, and saw a subsequent knockdown of LMO2 and MYBL1 at both the mRNA and protein level. We confirmed seed sequence specificity by comparing miR-223 knockdown of luciferase reporter activity on the LMO2 3′UTR compared to its seed sequence mutant. We further found that miR-9 and miR-30 family members directly regulate PRDM1 (blimp1), a master regulator of the GC to PC transition. In U266 cells (PC-derived), introduction of miR-9 and miR-30 family precursor resulted in decreased PRDM1 protein expression, although transcript levels were not changed, consistent with previous evidence that miRNA can regulate at the post-transcriptional steps. We further profiled over 50 tumors derived from various B cell malignancies (small lymphocytic lymphoma, Burkitt lymphoma, and the molecular subsets of diffuse large B cell lymphoma) and found that these malignancies maintain the expression patterns of their respective lineage; microRNA expression profiles of normal B cells could correctly classify the lineage of these tumors in over 80% of the cases. In contrast to other malignancies, common lymphomas do not down-regulate microRNAs, but rather maintain the microRNA-expression patterns of their normal B-cell counterparts. Conclusion: Through concomitant microRNA and mRNA-profiling, we demonstrate a regulatory role for microRNAs at every stage in mature B-cell differentiation. Further, we have experimentally identified a direct role for the microRNA-regulation of key transcription factors in B-cell differentiation: LMO2, MYBL1 and PRDM1 (Blimp1). Thus, our data demonstrate that microRNAs may be important in maintaining the mature B-cell phenotype in normal and malignant B-cells.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3536-3536 ◽  
Author(s):  
David Dominguez-Sola ◽  
Jennifer Kung ◽  
Victoria A Wells ◽  
Antony B Holmes ◽  
Laura Pasqualucci ◽  
...  

Abstract A significant fraction of B cell non-Hodgkin lymphomas (B-NHL) of germinal center origin carry heterozygous missense mutations in FOXO1, a member of the FOXO family of transcription factors. FOXO1 is a central component of the PI3K signaling cascade engaged by the B cell receptor and is essential for B cell homeostasis and survival (Dengler et al, Nat Immunol 2008; Srinivasan et al, Cell 2009; Lin et al, Nat Immunol 2010). In response to PI3K activation, AKT phosphorylates FOXO1 leading to its nuclear-cytoplasmic translocation and inactivation. Missense mutations of the FOXO1 gene are detectable in germinal center (GC)-derived B-NHL, including ~12% of Burkitt Lymphoma (BL) and ~9% of Diffuse Large B Cell Lymphoma (DLBCL) cases (Schmitz et al, Nature 2012; Trinh et al, Blood 2013; Pasqualucci et al, Cell Rep 2014). The role of FOXO1 in normal GC development as well as the contribution of its mutations to lymphomagenesis is unclear. We show that FOXO1 expression is restricted to the dark zone of GCs, where its nuclear localization is detectable in most B cells. Mice carrying the conditional inactivation of FOXO1 in GC B cells display normal GC in number and size. However, these GCs lack phenotypically defined (CXCR4hi/CD86lo) dark zones and are entirely composed by light zone B cells (CXCR4lo/CD86hi). FOXO1-/- GC B cells express AICDA and carry a normal number of mutations in their immunonoglobulin genes, but do not undergo affinity maturation, resulting in severely impaired antigen responses. In order to identify the biological program controlled by FOXO1 in GC B cells, we identified candidate transcriptional target genes by integrating ChIP-seq and gene expression data. These analyses showed that that the establishment of the dark zone fate relies on a FOXO1-dependent transcriptional network that is enriched for genes involved in immune signaling cascades triggered by the B cell receptor and by a variety of cytokines controlling GC polarity. Notably, a majority of these target genes are co-bound and co-regulated, in a FOXO1-dependent manner, by BCL6, a well characterized GC master regulator. To assess the role of BL- and DLBCL-associated mutations, we first investigated the subcellular localization of FOXO1 mutant proteins by transfecting wild type and mutant GFP-tagged FOXO1 alleles into HeLa cells. As previously shown (Trinh et al, Blood 2013), this analysis showed that mutant FOXO1 proteins, but not the wild-type one, readily localize in the nucleus. Analogously, immunofluorescence analysis of BL and DLBCL samples showed the presence of nuclear FOXO1 in all tumors carrying mutations in the FOXO1 gene. However, nuclear localization was also detectable in virtually all cases carrying normal FOXO1 genes. Accordingly, in vitro experiments testing the ability of normal and mutated FOXO1 proteins to respond to various signals activating the PI3K pathway in multiple BL and DLBCL cell lines, failed to display a correlation between the presence of mutations and responsiveness to these signals. Taken together, these results suggest that other mechanisms in addition to direct gene mutation are responsible for the constitutive nuclear localization of FOXO1 in tumors. We are now examining the consequences of FOXO1 missense mutations in vivo, by reconstituting FOXO1-/- GC B cells with FOXO1 mutants using bone marrow chimeras. Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 213 (7) ◽  
pp. 1201-1221 ◽  
Author(s):  
Miriam Wöhner ◽  
Hiromi Tagoh ◽  
Ivan Bilic ◽  
Markus Jaritz ◽  
Daniela Kostanova Poliakova ◽  
...  

E2A is an essential regulator of early B cell development. Here, we have demonstrated that E2A together with E2-2 controlled germinal center (GC) B cell and plasma cell development. As shown by the identification of regulated E2A,E2-2 target genes in activated B cells, these E-proteins directly activated genes with important functions in GC B cells and plasma cells by inducing and maintaining DNase I hypersensitive sites. Through binding to multiple enhancers in the Igh 3′ regulatory region and Aicda locus, E-proteins regulated class switch recombination by inducing both Igh germline transcription and AID expression. By regulating 3′ Igk and Igh enhancers and a distal element at the Prdm1 (Blimp1) locus, E-proteins contributed to Igk, Igh, and Prdm1 activation in plasmablasts. Together, these data identified E2A and E2-2 as central regulators of B cell immunity.


Sign in / Sign up

Export Citation Format

Share Document