A Critical BCL6-Related Feedback Loop Explains the Unusual Biological Features of Germinal Center B-Cells and Their Malignant Transformation into B-Cell Lymphomas.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 224-224 ◽  
Author(s):  
Stella M. Ranuncolo ◽  
Jose M. Polo ◽  
Jamil Dierov ◽  
Martin Carroll ◽  
Ari M. Melnick

Abstract The BCL6 (B-Cell-Lymphoma-6) transcriptional repressor is a critical oncogene in B-cell lymphomas and is required for establishment of germinal centers (GCs) by normal B-cells. However, the mechanisms by which BCL6 licenses GC formation and lymphomagenesis remain unknown. To characterize this mechanism we identified BCL6 target genes through several methods. Most notable among these was ATR, a master DNA damage response mediator. We showed that primary BCL6-expressing GC centroblasts purified from human tonsils do not express ATR, do not activate downstream targets of ATR (Chk1) and exhibit an overall attenuated DNA damage checkpoint response (as shown in COMET assays, H2AX phosphorylation assays, and other methods). ATR expression, the activation of ChK1, and the DNA damage phenotype were fully rescued by BCL6 loss of function (induced by shRNA or by a specific BCL6 inhibitor that we designed). BCL6 expressing DLBCL cell lines exhibited the same BCL6-dependent DNA damage response phenotype. This could be attributed almost entirely to ATR since ectopic expression of ATR could restore DNA damage sensing even in the presence of BCL6. Reciprocally, loss of ATR in BCL6 depleted B-cells could completely prevent them from restoring their damage response pathways. These effects are independent of p53, which in contrast to ATR is expressed in primary centroblasts and in many primary DLBCL cases. Gain of function experiments with BCL6 in isolated human tonsilar naïve B-cells could induce this same damage phenotype in an ATR dependent/p53 independent manner. Most remarkably, we discovered that CD40 signaling, which occurs once centroblasts mature into centrocytes in the GC light zone, rescues ATR from BCL6 mediated repression by signaling to the BCL6 repression complex through NFKB. This causes the SMRT and N-CoR corepressors to be released from BCL6 repressor complexes associated with the ATR promoter, induces ATR expression, its downstream target ChK1 activation and restores DNA damage sensing. As a consequence, B-cells that have undergone extensive damage (as a by-product of antibody affinity maturation and intense proliferation) can now undergo apoptosis, while those with lower level damage undergo cell cycle arrest, DNA repair, and further differentiation. We thus described a molecular feedback loop through which BCL6 mediates GC formation by directly repressing ATR and inducing a DNA damage checkpoint deficient phenotype, and through which subsequent CD40 signaling rescues this phenotype by disrupting the BCL6 repression complex. We showed that sustained BCL6 expression leads to DNA misrepair and genomic instability in primary B cells consistent with its role in lymphomagenesis. Reciprocally, therapeutic targeting of BCL6 synergized with chemotherapy and radiation to kill DLBCL cells (by restoring DNA damage checkpoints) and would thus be expected to improve therapeutic outcomes of DLBCL patients.

2019 ◽  
Vol 18 (7) ◽  
pp. 1255-1264 ◽  
Author(s):  
Valentina Restelli ◽  
Monica Lupi ◽  
Rosaria Chilà ◽  
Micaela Vagni ◽  
Chiara Tarantelli ◽  
...  

Blood ◽  
2013 ◽  
Vol 121 (15) ◽  
pp. 2964-2974 ◽  
Author(s):  
Jake Shortt ◽  
Benjamin P. Martin ◽  
Andrea Newbold ◽  
Katherine M. Hannan ◽  
Jennifer R. Devlin ◽  
...  

Key Points MYC-driven lymphomas demonstrate activation of mTORC1 and an endogenous DNA damage response. BEZ235 inhibits PI3K-related DNA damage response kinases and mTORC1, inducing p53-independent upregulation of proapoptotic BMF.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2279-2279
Author(s):  
Hakim Bouamar ◽  
Long Wang ◽  
Manoela Ortega ◽  
An-Ping Lin ◽  
Daifeng Jiang ◽  
...  

Abstract The germinal center (GC) reaction includes two critical events: somatic hypermutation (SHM) and class switch DNA recombination (CSR). These processes are tightly controlled, thus preventing excessive DNA injury which could lead to loss of normal B lymphocytes as well as the survival of cells with oncogenic DNA lesions. MicroRNA-155 (miR-155) plays an important role in immune cell biology; miR-155 knock-out (KO) mice have a defective mature B cell development characterized by a decreased number of GC B cells, whereas the Eμ-miR-155 transgenic mouse model develops and oligoclonal proliferation which evolves to B cell lymphoma. These observations suggest that miR-155 may regulate B cell sensitivity and response to DNA damage, which could mechanistically explain the phenotypes observed in the gain and loss-of-function animal models. To examine this concept, we purified mature B cells from multiple pairs of miR-155 KO and WT littermates, and stimulated them with LPS and IL4 thus recapitulating the GC reaction in vitro. Next, we used immunohistochemistry to quantify γH2AX at the DNA double-strand breaks (DSBs) foci, western blot to evaluate the phospho-p53 (Ser15) levels, and real-time RT-PCR to quantify the expression of p53 target genes (p21, GADD45a, cdc25c, PCNA). In these assays, we found a significantly higher γH2AX staining in B cells null for miR-155 than in WT controls (number of foci per nucleus: 4.1±0.9 vs. 1.4±0.2, p<0.01), and a markedly elevated p53 activity, defined by its phospho-level and target genes expression. To confirm these observations in a more physiologic setting, we immunized miR-155 WT and KO mice with NP-CGG, collected spleens and purified mature B-cells. Quantification of subpopulation by FACS confirmed that miR-155 mice developed fewer GC B cells, and the examination of DSBs foci and p53 target genes expression supported our in vitro observation of a heightened sensitivity to DNA damage and p53 engagement in miR-155 null cells. We concluded that miR-155 deficiency in B cells associates with excessive DNA damage and p53 activity. To further isolate define the role of miR-155 in these events, we used a retrovirus system to rescue miR-155 expression in B cells derived from the KO mouse, and defined the pattern of DNA damage response. In these assays, cells transduced with a MSCV-miR-155 construct formed fewer DSBs foci than their control counterparts transduced with an empty vector (9.0±2.1 vs. 4.9±1.8, p<0.05) and showed significantly lower p53 activity, defined by target gene expression. To test if miR-155 controls the DNA damage induced by broad genotoxic stresses, we exposed miR-155 KO or WT B cells, thymocytes and bone marrow cells to 5Gy of ionizing radiation (IR) or etoposide (4µM). In these assays, albeit a robust induction of γH2AX foci formation and p53 activation were detected, there was no significant difference between WT and miR-155 KO mice, in any of the cell types analyzed. We concluded that the heightened sensitivity to genotoxic stress in miR-155 KO mice is specific to B cells undergoing the GC reaction. This observation suggests a potential role in this process for activation-induced cytidine deaminase (AID), a key regulator of the DNA damage inducing SHM and CSR, which is also a miR-155 target gene. We confirmed that AID expression is higher in miR-155 KO than in WT B cells, and to test its contribution to the excessive DSB and p53 activity found miR-155 null cells we used an RNAi approach. We found that the inhibition of AID levels in miR-155 KO B cells significantly reduced γH2AX foci formation and expression of p53 target genes. Together, our data highlight a hitherto unappreciated interaction between miR-155 and the p53 pathway, involving DNA lesions that are germane to the GC reaction and the control of AID expression/activity. The excessive engagement of p53 is this setting may explain, at least in part, the loss of normal GC B cells found in the miR-155 KO mice. Conversely, it is possible that cells expressing abnormally high miR-155 levels show a subpar activation of the DNA damage response thus leading to the accumulation of oncogenic mutations. This paradigm would provide a mechanistic explanation for the initial poly/oligoclonal proliferation reported in the Eμ-miR-155 mice, which eventually evolves into a B-cell lymphoma Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 157-157 ◽  
Author(s):  
Stella M. Ranuncolo ◽  
Jose M. Polo ◽  
Jamil Dierov ◽  
Rita Shaknovich ◽  
Martin Carroll ◽  
...  

Abstract The BCL6 (B-Cell-Lymphoma-6) transcriptional repressor is a critical oncogene in B-cell lymphomas and is required for establishment of germinal centers by normal B-cells. However, the mechanisms by which BCL6 licenses germinal center formation and lymphomagenesis are unknown. To characterize this mechanism we identified BCL6 target genes by expression arrays and high throughput chromatin immunoprecipitations. Remarkably, a number of these target genes were critical mediators of DNA damage sensing checkpoints including ATR and p53. Therefore, we hypothesized that BCL6 could attenuate DNA damage sensing by silencing these genes, which is likely a critical attribute for survival and proliferation of germinal center B-cells cells undergoing somatic hypermutation (SHM) and class switch recombination (CSR). Accordingly, we found that expression of BCL6 in normal diploid fibroblasts could block cellular sensing of DNA damage as demonstrated by loss of histone 2AX (H2AX) phosphorylation and delayed repair of double strand breaks. Repression of ATR (but not p53 or other targets) was required for this phenotype. This is a physiological effect since the same result was observed when BCL6 was expressed in purified primary human tonsilar mature B-cells. Reciprocally, shRNA knockdown of BCL6 in B-cell lymphomas rescued repression of ATR, enhanced H2AX phosphorylation and accelerated repair of double strand breaks, independent of the status of p53. shRNA knockdown of BCL6 caused a marked increase of apoptosis in lymphoma cells in response to DNA damage, due to restored DNA damage checkpoint functions. Importantly, BCL6 knockdown had an identical effect on ATR levels, H2AX phosphorylation, DNA damage, and survival in purified primary human germinal center centroblasts. These results suggest that a major role of BCL6 in germinal center formation is to attenuate cellular response to DNA damage occurring as a byproduct of CSR and SHM. The same mechanism also seems to be required for lymphomagenesis, since we observed that sustained BCL6 expression in human primary mature B-cells leads to aberrant survival properties and genomic instability. Moreover, BCL6 blockade using a specific inhibitor molecule designed by our lab induces apoptosis in lymphoma cells and synergizes with DNA damaging agents. Therefore, we have identified a critical mechanism of action of the BCL6 oncoprotein in normal and pathogenic states and show that specific targeting of BCL6 could synergize with chemotherapy drugs for the therapy of B-cell lymphomas.


2006 ◽  
Vol 17 (1) ◽  
pp. 539-548 ◽  
Author(s):  
Tania M. Roberts ◽  
Michael S. Kobor ◽  
Suzanne A. Bastin-Shanower ◽  
Miki Ii ◽  
Sonja A. Horte ◽  
...  

RTT107 (ESC4, YHR154W) encodes a BRCA1 C-terminal-domain protein that is important for recovery from DNA damage during S phase. Rtt107 is a substrate of the checkpoint protein kinase Mec1, although the mechanism by which Rtt107 is targeted by Mec1 after checkpoint activation is currently unclear. Slx4, a component of the Slx1-Slx4 structure-specific nuclease, formed a complex with Rtt107. Deletion of SLX4 conferred many of the same DNA-repair defects observed in rtt107Δ, including DNA damage sensitivity, prolonged DNA damage checkpoint activation, and increased spontaneous DNA damage. These phenotypes were not shared by the Slx4 binding partner Slx1, suggesting that the functions of the Slx4 and Slx1 proteins in the DNA damage response were not identical. Of particular interest, Slx4, but not Slx1, was required for phosphorylation of Rtt107 by Mec1 in vivo, indicating that Slx4 was a mediator of DNA damage-dependent phosphorylation of the checkpoint effector Rtt107. We propose that Slx4 has roles in the DNA damage response that are distinct from the function of Slx1-Slx4 in maintaining rDNA structure and that Slx4-dependent phosphorylation of Rtt107 by Mec1 is critical for replication restart after alkylation damage.


Blood ◽  
2021 ◽  
Author(s):  
Miguel A Galindo-Campos ◽  
Nura Lutfi ◽  
Sarah Bonnin ◽  
Carlos Martínez ◽  
Talia Velasco-Hernandez ◽  
...  

Dysregulation of the c-Myc oncogene occurs in a wide variety of haematologic malignancies and its overexpression has been linked with aggressive tumour progression. Here, we show that Poly (ADP-ribose) polymerase (PARP)-1 and PARP-2 exert opposing influences on progression of c-Myc-driven B-cell lymphomas. PARP-1 and PARP-2 catalyse the synthesis and transfer of ADP-ribose units onto amino acid residues of acceptor proteins in response to DNA-strand breaks, playing a central role in the response to DNA damage. Accordingly, PARP inhibitors have emerged as promising new cancer therapeutics. However, the inhibitors currently available for clinical use are not able to discriminate between individual PARP proteins. We found that genetic deletion of PARP-2 prevents c-Myc-driven B-cell lymphomas, while PARP-1-deficiency accelerates lymphomagenesis in the Em-Myc mouse model of aggressive B-cell lymphoma. Loss of PARP-2 aggravates replication stress in pre-leukemic Em-Myc B cells resulting in accumulation of DNA damage and concomitant cell death that restricts the c-Myc-driven expansion of B cells, thereby providing protection against B-cell lymphoma. In contrast, PARP-1-deficiency induces a proinflammatory response, and an increase in regulatory T cells likely contributing to immune escape of B-cell lymphomas, resulting in an acceleration of lymphomagenesis. These findings pinpoint specific functions for PARP-1 and PARP-2 in c-Myc-driven lymphomagenesis with antagonistic consequences that may help inform the design of new PARP-centred therapeutic strategies with selective PARP-2 inhibition potentially representing a new therapeutic approach for the treatment of c-Myc-driven tumours.


2021 ◽  
Vol 31 (02) ◽  
pp. 2150020
Author(s):  
Chunyan Gao ◽  
Fangqi Chen

This study develops a general model of delayed p53 regulatory network in the DNA damage response by introducing microRNA 192-mediated positive feedback loop based on the existing research work. Through theoretical analysis and numerical simulation, we find that the delay as a bifurcation parameter can drive the p53-Mdm2 module to undergo a supercritical Hopf bifurcation, thereby producing oscillation behavior. Moreover, we demonstrate how the positive feedback loop formed by p53* and microRNA 192 (miR-192) with the feature of double-negative regulation produces oscillations. Further, a comparison is given to demonstrate that microRNA 192-mediated positive feedback loop affects the robustness of system oscillations. In addition, we show that ataxia telangiectasia mutated kinase (ATM), once activated by DNA damage, makes p53* undergo two Hopf bifurcations. These results reveal that both time delay and miR-192 play tumor suppressing roles by promoting p53 oscillation or high level expression, which will provide a perspective for promoting the development of anti-cancer drugs by targeting miR-192 and time delay.


Genetics ◽  
2021 ◽  
Author(s):  
Tingting Li ◽  
Ruben C Petreaca ◽  
Susan L Forsburg

Abstract Chromatin remodeling is essential for effective repair of a DNA double strand break. KAT5 (S. pombe Mst1, human TIP60) is a MYST family histone acetyltransferase conserved from yeast to humans that coordinates various DNA damage response activities at a DNA double strand break (DSB), including histone remodeling and activation of the DNA damage checkpoint. In S. pombe, mutations in mst1+ causes sensitivity to DNA damaging drugs. Here we show that Mst1 is recruited to DSBs. Mutation of mst1+ disrupts recruitment of repair proteins and delays resection. These defects are partially rescued by deletion of pku70, which has been previously shown to antagonize repair by homologous recombination. These phenotypes of mst1 are similar to pht1-4KR, a non-acetylatable form of histone variant H2A.Z, which has been proposed to affect resection. Our data suggest that Mst1 functions to direct repair of DSBs towards homologous recombination pathways by modulating resection at the double strand break.


2018 ◽  
Vol 9 (5) ◽  
Author(s):  
Rongfu Tu ◽  
Wenqian Kang ◽  
Xuefei Yang ◽  
Qi Zhang ◽  
Xiaoyu Xie ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document