scholarly journals Multi-tiered Reorganization of the Genome during B Cell Affinity Maturation Anchored by a Germinal Center-Specific Locus Control Region

Immunity ◽  
2016 ◽  
Vol 45 (3) ◽  
pp. 497-512 ◽  
Author(s):  
Karen L. Bunting ◽  
T. David Soong ◽  
Rajat Singh ◽  
Yanwen Jiang ◽  
Wendy Béguelin ◽  
...  
Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 24-24
Author(s):  
Johannes C Hellmuth ◽  
Chi-Shuen Chu ◽  
Rajat Singh ◽  
Lucy A Skrabanek ◽  
Ashley Doane ◽  
...  

The transcriptional repressor BCL6 is considered the master regulator of the germinal center (GC) reaction and is a key proto-oncogene in GC-derived lymphoma pathogenesis. At GC initiation, chromatin architecture is dramatically remodeled around a BCL6-associated locus control region (LCR) with presumed enhancer function. The BCL6 LCR is located 150kb upstream of the BCL6 gene and is fundamentally required for GC formation. Herein, we functionally dissect the BCL6 LCR to uncover the crucial genetic elements and transcription factor binding events that drive BCL6 LCR function. We generated a custom gRNA library densely tiling the BCL6 LCR and surrounding regulatory elements. In total, the library covered a genomic region of 316.8kb with 25,698 gRNAs. We used a GC derived diffuse large B-cell lymphoma line, OCI-LY7, stably expressing dCas9-KRAB (CRISPRi) to screen for depletion of gRNAs. Surprisingly, BCL6 LCR function relied on only 4 of 21 constituent enhancers. These 4 essential enhancers were critically required for cell growth and BCL6 expression. In contrast, all other constituent enhancers were completely dispensable for LCR function. These results indicate that the BCL6 LCR is governed by a strong internal hierarchy and pinpoint the crucial genetic elements that drive LCR function. To understand what distinguishes essential enhancers from non-essential enhancers, we interrogated ChIP-seq profiles of key GC transcription factors (TF) and transcriptional co-activators. While most TFs and co-activators bound constituent enhancers indiscriminately, MEF2B specifically bound only to essential enhancers (p=0.01). Interestingly, essential enhancers did not contain MEF2B binding motifs. Furthermore, MEF2B did not bind to essential enhancer DNA in electrophoretic mobility shift (EMSA) and immobilized template assays. De novo motif analysis of MEF2B ChIP-seq data prominently featured the canonical octamer transcription factor motif (p=10-79) indicating a role for OCT2 in the recruitment of MEF2B to essential enhancers. Indeed, MEF2B binding to essential enhancer DNA in EMSA and immobilized template assays was dependent upon the presence of OCT2 and its co-activator OCA-B. We assayed occupancy of OCT2, OCA-B and MEF2B at essential enhancers in OCI-LY7 cells by qChIP and found that each factor required the other two for full binding activity. Furthermore, all three factors were required for BCL6 expression (p<0.001 for each factor). These results indicate that OCT2, OCA-B and MEF2B cooperatively bind to essential enhancer elements and act as an intimately linked ternary complex to drive BCL6 expression through the BCL6 LCR. To elucidate how the OCT2 / OCA-B / MEF2B complex promotes target gene transcription, we performed IP of OCA-B followed by mass spectrometry and found highly significant interactions with the majority of Mediator proteins (p<10-8). We furthermore showed that OCA-B directly and specifically interacts with MED1, through which it recruits the remainder of the Mediator complex and that OCA-B and OCT2 are required to recruit Mediator to essential enhancer elements. The Mediator complex is thought to serve as a drawbridge across enhancers and promoters to facilitate enhancer-promoter looping. Using 3C assays, we found that OCA-B is required for chromatin contacts between the BCL6 promoter and the LCR highlighting its importance in recruiting Mediator. Similarly, essential enhancer elements were crucially required for intact chromatin conformation at the BCL6 locus as determined by 3C. In summary, BCL6 LCR function completely relies on very few but highly essential enhancer elements. OCT2, OCA-B and MEF2B cooperatively bind these essential enhancers forming an intimately linked trimeric complex. By recruiting Mediator, OCA-B provides a direct link to the basal transcriptional machinery. Finally, essential enhancers as well as the OCT2 / OCA-B / MEF2B complex are required for BCL6 expression and intact chromatin conformation at the BCL6 locus - key determinants of the GC B cell state. Disclosures Melnick: Epizyme: Consultancy; Constellation: Consultancy; Janssen: Research Funding.


2020 ◽  
Vol 217 (9) ◽  
Author(s):  
Xin Li ◽  
Liying Gong ◽  
Alexandre P. Meli ◽  
Danielle Karo-Atar ◽  
Weili Sun ◽  
...  

Antigen uptake and presentation by naive and germinal center (GC) B cells are different, with the former expressing even low-affinity BCRs efficiently capture and present sufficient antigen to T cells, whereas the latter do so more efficiently after acquiring high-affinity BCRs. We show here that antigen uptake and processing by naive but not GC B cells depend on Cbl and Cbl-b (Cbls), which consequently control naive B and cognate T follicular helper (Tfh) cell interaction and initiation of the GC reaction. Cbls mediate CD79A and CD79B ubiquitination, which is required for BCR-mediated antigen endocytosis and postendocytic sorting to lysosomes, respectively. Blockade of CD79A or CD79B ubiquitination or Cbls ligase activity is sufficient to impede BCR-mediated antigen processing and GC development. Thus, Cbls act at the entry checkpoint of the GC reaction by promoting naive B cell antigen presentation. This regulation may facilitate recruitment of naive B cells with a low-affinity BCR into GCs to initiate the process of affinity maturation.


Author(s):  
Yanan Li ◽  
Anshuman Bhanja ◽  
Arpita Upadhyaya ◽  
Xiaodong Zhao ◽  
Wenxia Song

B-cells undergo somatic hypermutation and affinity maturation in germinal centers. Somatic hypermutated germinal center B-cells (GCBs) compete to engage with and capture antigens on follicular dendritic cells. Recent studies show that when encountering membrane antigens, GCBs generate actin-rich pod-like structures with B-cell receptor (BCR) microclusters to facilitate affinity discrimination. While deficiencies in actin regulators, including the Wiskott-Aldrich syndrome protein (WASp), cause B-cell affinity maturation defects, the mechanism by which actin regulates BCR signaling in GBCs is not fully understood. Using WASp knockout (WKO) mice that express Lifeact-GFP and live-cell total internal reflection fluorescence imaging, this study examined the role of WASp-mediated branched actin polymerization in the GCB immunological synapse. After rapid spreading on antigen-coated planar lipid bilayers, GCBs formed microclusters of phosphorylated BCRs and proximal signaling molecules at the center and the outer edge of the contact zone. The centralized signaling clusters localized at actin-rich GCB membrane protrusions. WKO reduced the centralized micro-signaling clusters by decreasing the number and stability of F-actin foci supporting GCB membrane protrusions. The actin structures that support the spreading membrane also appeared less frequently and regularly in WKO than in WT GCBs, which led to reductions in both the level and rate of GCB spreading and antigen gathering. Our results reveal essential roles for WASp in the generation and maintenance of unique structures for GCB immunological synapses.


2020 ◽  
Author(s):  
Juhee Pae ◽  
Jonatan Ersching ◽  
Tiago B. R. Castro ◽  
Marta Schips ◽  
Luka Mesin ◽  
...  

AbstractDuring affinity maturation, germinal center (GC) B cells alternate between proliferation and so-matic hypermutation in the dark zone (DZ) and affinity-dependent selection in the light zone (LZ). This anatomical segregation imposes that the vigorous proliferation that allows clonal expansion of positively-selected GC B cells takes place ostensibly in the absence of the signals that triggered selection in the LZ, as if by “inertia.” We find that such inertial cycles specifically require the cell cycle regulator cyclin D3. Cyclin D3 dose-dependently controls the extent to which B cells proliferate in the DZ and is essential for effective clonal expansion of GC B cells in response to strong T follicular helper (Tfh) cell help. Introduction into the Ccnd3 gene of a Burkitt lymphoma-associated gain-of-function mutation (T283A) leads to larger GCs with increased DZ proliferation and, in older mice, to clonal B cell lymphoproliferation, suggesting that the DZ inertial cell cycle program can be coopted by B cells undergoing malignant transformation.


2018 ◽  
Vol 215 (6) ◽  
pp. 1571-1588 ◽  
Author(s):  
Norbert Pardi ◽  
Michael J. Hogan ◽  
Martin S. Naradikian ◽  
Kaela Parkhouse ◽  
Derek W. Cain ◽  
...  

T follicular helper (Tfh) cells are required to develop germinal center (GC) responses and drive immunoglobulin class switch, affinity maturation, and long-term B cell memory. In this study, we characterize a recently developed vaccine platform, nucleoside-modified, purified mRNA encapsulated in lipid nanoparticles (mRNA-LNPs), that induces high levels of Tfh and GC B cells. Intradermal vaccination with nucleoside-modified mRNA-LNPs encoding various viral surface antigens elicited polyfunctional, antigen-specific, CD4+ T cell responses and potent neutralizing antibody responses in mice and nonhuman primates. Importantly, the strong antigen-specific Tfh cell response and high numbers of GC B cells and plasma cells were associated with long-lived and high-affinity neutralizing antibodies and durable protection. Comparative studies demonstrated that nucleoside-modified mRNA-LNP vaccines outperformed adjuvanted protein and inactivated virus vaccines and pathogen infection. The incorporation of noninflammatory, modified nucleosides in the mRNA is required for the production of large amounts of antigen and for robust immune responses.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 279-279
Author(s):  
Karen L. Bunting ◽  
David Soong ◽  
Yanwen Jiang ◽  
Brandon L. Swed ◽  
Katerina Hatzi ◽  
...  

Abstract Abstract 279 B cell affinity maturation is fundamental to the development of humoral immunity. To create a diverse antibody repertoire, B cells activated in the germinal centre (GC) must undergo a profound change in phenotype. This unique phenotypic change, which features simultaneous proliferation and somatic hypermutation and which can predispose to the development of lymphoma, requires radically altered gene expression programming in GC B cells. However, the way that this gene expression program is coordinated is unknown. Emerging evidence suggests that the higher-order organization of chromatin plays a role in the co-regulation of genes. We hypothesised that the three-dimensional organization of genes and chromosomes in the nucleus of B cells plays a key role in the epigenetic and transcriptional reprogramming that underlies acquisition of the GC B cell phenotype during B cell maturation. Using genome-wide mapping of chromatin interactions (Hi-C), combined with genome-wide profiles of gene expression (RNA-seq), histone modifications and transcription factor binding (ChIP-seq) in human naïve B (NB) and GC B cells, we have discovered that the three-dimensional structure of the genome undergoes widespread reorganization during B cell maturation to coordinate the GC transcriptional programme. Conformational maps of chromosome folding in these cells reveal a novel and profound loss of inter-arm interactions, reflecting lower chromosome compaction in GC B cells. Remarkably, we observed extensive differential partitioning of genes into NB- and GC B cell-specific compartments, and demonstrate for the first time that coordinated changes in histone modifications (H3K4Me2: P=3×10−35; H3K27Ac: P=3×10−33; Fisher's exact test) and transcription (P=1×10−9) required for cell type specification is mediated by the de novo formation of precisely delimited chromosome neighbourhoods. Most strikingly, we find that remodelling of the GC B cell genome involves the specific structural unlocking of genes that drive the GC transcriptional programme, such as AICDA, MTA3, and BCL6. Coordinate activation of these genes is mediated by the expansion of gene interaction neighbourhoods, increased promoter interactivity (P=3×10−35), engagement of long-range enhancer-promoter interactions (>2-fold increase), and the formation of gene body loops (P=3.18×10−15). Intriguingly, the master regulator of GC B cell differentiation, BCL6, shows a high propensity for all of these different types of interactions, suggesting that regulation of this gene in the context of chromatin is highly complex. Integration with genome-wide binding data for the structural organizing proteins, CTCF and cohesin, as well as the cell-specific factor, PU.1, supports a specific role for these proteins in the repositioning of activated promoters and enhancer regions during B cell maturation. This study shows for the first time that the architecture of the genome is critical for specification of cellular phenotype, and that epigenetic and transcriptional reprogramming in GC B cells is functionally linked to the structural reorganization of genes in the nucleus. Importantly, the higher-order organization of chromatin could represent a novel mechanism by which GC B cell gene expression is dysregulated in lymphoma. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document